1
|
Dadson P, Honka MJ, Suomi T, Haridas PAN, Rokka A, Palani S, Goltseva E, Wang N, Roivainen A, Salminen P, James P, Olkkonen VM, Elo LL, Nuutila P. Proteomic profiling reveals alterations in metabolic and cellular pathways in severe obesity and following metabolic bariatric surgery. Am J Physiol Endocrinol Metab 2025; 328:E311-E324. [PMID: 39819027 DOI: 10.1152/ajpendo.00220.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
In this study, we investigated the impact of bariatric surgery on the adipose proteome to better understand the metabolic and cellular mechanisms underlying weight loss following the procedure. A total of 46 patients with severe obesity were included, with samples collected both before and after bariatric surgery. In addition, 15 healthy individuals without obesity who did not undergo surgery served as controls and were studied once. We utilized quantitative liquid chromatography-tandem mass spectrometry analysis to conduct a large-scale proteomic study on abdominal subcutaneous biopsies obtained from the study participants. Our proteomic profiling revealed that among the 2,254 compared proteins, 46 were upregulated and 34 were downregulated 6 months post surgery compared with baseline [false discovery rate (FDR) < 0.01]. We observed a downregulation of proteins associated with mitochondrial integrity, amino acid catabolism, and lipid metabolism in the patients with severe obesity compared with the controls. Bariatric surgery was associated with an upregulation in pathways related to mitochondrial function, protein synthesis, folding and trafficking, actin cytoskeleton regulation, and DNA binding and repair. These findings emphasize the significant changes in metabolic and cellular pathways following bariatric surgery, highlighting the potential mechanisms underlying the observed health improvements postbariatric surgery. The data provided alongside this paper will serve as a valuable resource for the development of targeted therapeutic strategies for obesity and related metabolic complications. ClinicalTrials.gov registration numbers: NCT00793143 (registered on 19 November 2008) (https://clinicaltrials.gov/ct2/show/NCT00793143) and NCT01373892 (registered on 15 June 2011) (https://clinicaltrials.gov/ct2/show/NCT01373892).NEW & NOTEWORTHY Our study investigates the effects of metabolic bariatric surgery on adipose tissue proteins, highlighting the mechanisms driving weight loss postsurgery. Through extensive proteomic analysis of adipose biopsies from patients with severe obesity pre- and postsurgery, alongside healthy subjects without obesity, we identified significant alterations in metabolic pathways. These findings provide insights into potential therapeutic targets for obesity-related complications.
Collapse
Affiliation(s)
- Prince Dadson
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Miikka-Juhani Honka
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Division of Information Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Anne Rokka
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Elena Goltseva
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Ning Wang
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Paulina Salminen
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Department of Surgery, University of Turku, Turku, Finland
- Division of Digestive Surgery and Urology, Turku University Hospital, Turku, Finland
| | - Peter James
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| |
Collapse
|
2
|
Deng WQ, Ye ZH, Tang Z, Zhang XL, Lu JJ. Beyond cancer: The potential application of CD47-based therapy in non-cancer diseases. Acta Pharm Sin B 2025; 15:757-791. [PMID: 40177549 PMCID: PMC11959971 DOI: 10.1016/j.apsb.2024.11.018] [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: 08/29/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
CD47 is an immune checkpoint widely regarded as a 'don't eat me' signal. CD47-based anti-cancer therapy has received considerable attention, with a significant number of clinical trials conducted. While anti-cancer therapies based on CD47 remain a focal point of interest among researchers, it is noteworthy that an increasing number of studies have found that CD47-based therapy ameliorated the pathological status of non-cancer diseases. This review aims to provide an overview of the recent progress in comprehending the role of CD47-based therapy in non-cancer diseases, including diseases of the circulatory system, nervous system, digestive system, and so on. Furthermore, we sought to delineate the promising mechanisms of CD47-based therapy in treating non-cancer diseases. Our findings suggest that CD47-based agents may exert their effect by regulating phagocytosis, regulating T cells, dendritic cells, and neutrophils, and regulating the secretion of cytokines and chemokines. Additionally, we put forward the orientation of further research to bring to light the potential of CD47 and its binding partners as a target in non-cancer diseases.
Collapse
Affiliation(s)
- Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Zhenghai Tang
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Xiao-Lei Zhang
- National-Local Joint Engineering Laboratory of Druggability and New Drug Evaluation, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
3
|
Polara R, Ganesan R, Pitson SM, Robinson N. Cell autonomous functions of CD47 in regulating cellular plasticity and metabolic plasticity. Cell Death Differ 2024; 31:1255-1266. [PMID: 39039207 PMCID: PMC11445524 DOI: 10.1038/s41418-024-01347-w] [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: 03/07/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024] Open
Abstract
CD47 is a ubiquitously expressed cell surface receptor, which is widely known for preventing macrophage-mediated phagocytosis by interacting with signal regulatory protein α (SIRPα) on the surface of macrophages. In addition to its role in phagocytosis, emerging studies have reported numerous noncanonical functions of CD47 that include regulation of various cellular processes such as proliferation, migration, apoptosis, differentiation, stress responses, and metabolism. Despite lacking an extensive cytoplasmic signaling domain, CD47 binds to several cytoplasmic proteins, particularly upon engaging with its secreted matricellular ligand, thrombospondin 1. Indeed, the regulatory functions of CD47 are greatly influenced by its interacting partners. These interactions are often cell- and context-specific, adding a further level of complexity. This review addresses the downstream cell-intrinsic signaling pathways regulated by CD47 in various cell types and environments. Some of the key pathways modulated by this receptor include the PI3K/AKT, MAPK/ERK, and nitric oxide signaling pathways, as well as those implicated in glucose, lipid, and mitochondrial metabolism. These pathways play vital roles in maintaining tissue homeostasis, highlighting the importance of understanding the phagocytosis-independent functions of CD47. Given that CD47 expression is dysregulated in a variety of cancers, improving our understanding of the cell-intrinsic signals regulated by this molecule will help advance the development of CD47-targeted therapies.
Collapse
Affiliation(s)
- Ruhi Polara
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Raja Ganesan
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
- Institute for Molecular Immunology, CECAD Research Center, University Hospital Cologne, Cologne, Germany
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Nirmal Robinson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
4
|
Li Y, Dong L, Yin X, Wang X, Zhu X, Zheng P, Tang Y. CD47, a novel YAP target gene, contributes to hepatic stellate cell activation and liver fibrosis induced by high-fat diet. Heliyon 2024; 10:e31621. [PMID: 38831842 PMCID: PMC11145538 DOI: 10.1016/j.heliyon.2024.e31621] [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: 12/18/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
Activated hepatic stellate cells (HSCs) have been widely recognized as a primary source of pathological myofibroblasts, leading to the accumulation of extracellular matrix and liver fibrosis. CD47, a transmembrane glycoprotein expressed on the surface of various cell types, has been implicated in non-alcoholic fatty liver disease. However, the precise role of CD47 in HSC activation and the underlying regulatory mechanisms governing CD47 expression remain poorly understood. In this study, we employed single-cell RNA sequencing analysis to investigate CD47 expression in HSCs from mice subjected to a high-fat diet. CD47 silencing in HSCs markedly inhibited the expression of fibrotic genes and promoted apoptosis. Mechanistically, we found that Yes-associated protein (YAP) collaborates with TEAD4 to augment the transcriptional activation of CD47 by binding to its promoter region. Notably, disruption of the interaction between YAP and TEAD4 caused a substantial decrease in CD47 expression in HSCs and reduced the development of high-fat diet-induced liver fibrosis. Our findings highlight CD47 as a critical transcriptional target of YAP in promoting HSC activation in response to a high-fat diet. Targeting the YAP/TEAD4/CD47 signaling axis may hold promise as a therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Ya Li
- Department of Pediatrics, Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury and Outstanding Foreign Scientists Studio for Chronic Liver Injury, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Dong
- Department of Pediatrics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuecui Yin
- Department of Internal Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohan Wang
- Department of Pediatrics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohui Zhu
- Department of Pediatrics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Youcai Tang
- Department of Pediatrics, Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury and Outstanding Foreign Scientists Studio for Chronic Liver Injury, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Isenberg JS, Montero E. Tolerating CD47. Clin Transl Med 2024; 14:e1584. [PMID: 38362603 PMCID: PMC10870051 DOI: 10.1002/ctm2.1584] [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: 11/19/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Cluster of differentiation 47 (CD47) occupies the outer membrane of human cells, where it binds to soluble and cell surface receptors on the same and other cells, sculpting their topography and resulting in a pleiotropic receptor-multiligand interaction network. It is a focus of drug development to temper and accentuate CD47-driven immune cell liaisons, although consideration of on-target CD47 effects remain neglected. And yet, a late clinical trial of a CD47-blocking antibody was discontinued, existent trials were restrained, and development of CD47-targeting agents halted by some pharmaceutical companies. At this point, if CD47 can be exploited for clinical advantage remains to be determined. Herein an airing is made of the seemingly conflicting actions of CD47 that reflect its position as a junction connecting receptors and signalling pathways that impact numerous human cell types. Prospects of CD47 boosting and blocking are considered along with potential therapeutic implications for autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Jeffrey S. Isenberg
- Department of Diabetes Complications & MetabolismArthur Riggs Diabetes & Metabolism Research InstituteCity of Hope National Medical CenterDuarteCaliforniaUSA
| | - Enrique Montero
- Department of Molecular & Cellular EndocrinologyArthur Riggs Diabetes & Metabolism Research InstituteCity of Hope National Medical CenterDuarteCaliforniaUSA
| |
Collapse
|
6
|
Ghimire K, Kale A, Li J, Julovi SM, O'Connell P, Grey ST, Hawthorne WJ, Gunton JE, Rogers NM. A metabolic role for CD47 in pancreatic β cell insulin secretion and islet transplant outcomes. Sci Transl Med 2023; 15:eadd2387. [PMID: 37820008 DOI: 10.1126/scitranslmed.add2387] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/18/2023] [Indexed: 10/13/2023]
Abstract
Diabetes is a global public health burden and is characterized clinically by relative or absolute insulin deficiency. Therapeutic agents that stimulate insulin secretion and improve insulin sensitivity are in high demand as treatment options. CD47 is a cell surface glycoprotein implicated in multiple cellular functions including recognition of self, angiogenesis, and nitric oxide signaling; however, its role in the regulation of insulin secretion remains unknown. Here, we demonstrate that CD47 receptor signaling inhibits insulin release from human as well as mouse pancreatic β cells and that it can be pharmacologically exploited to boost insulin secretion in both models. CD47 depletion stimulated insulin granule exocytosis via activation of the Rho GTPase Cdc42 in β cells and improved glucose clearance and insulin sensitivity in vivo. CD47 blockade enhanced syngeneic islet transplantation efficiency and expedited the return to euglycemia in streptozotocin-induced diabetic mice. Further, anti-CD47 antibody treatment delayed the onset of diabetes in nonobese diabetic (NOD) mice and protected them from overt diabetes. Our findings identify CD47 as a regulator of insulin secretion, and its manipulation in β cells offers a therapeutic opportunity for diabetes and islet transplantation by correcting insulin deficiency.
Collapse
Affiliation(s)
- Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research (WIMR), University of Sydney, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145, Australia
| | - Atharva Kale
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research (WIMR), University of Sydney, Sydney, NSW 2145, Australia
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research (WIMR), University of Sydney, Sydney, NSW 2145, Australia
| | - Sohel M Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research (WIMR), University of Sydney, Sydney, NSW 2145, Australia
| | - Philip O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research (WIMR), University of Sydney, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145, Australia
| | - Shane T Grey
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Wayne J Hawthorne
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research (WIMR), University of Sydney, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145, Australia
| | - Jenny E Gunton
- Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145, Australia
- Centre for Diabetes, Obesity and Endocrinology, WIMR, University of Sydney, Sydney, NSW 2145, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research (WIMR), University of Sydney, Sydney, NSW 2145, Australia
- Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2145, Australia
| |
Collapse
|
7
|
Li D, Gwag T, Wang S. Sex differences in the effects of brown adipocyte CD47 deficiency on age-related weight change and glucose homeostasis. Biochem Biophys Res Commun 2023; 676:78-83. [PMID: 37499367 PMCID: PMC10810338 DOI: 10.1016/j.bbrc.2023.07.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
Our previous studies demonstrated that mice with global CD47 deficiency are lean and resistant to diet or aging-associated obesity and metabolic complications. This protective effect is partially through modulating brown fat function. To definitively determine the role of brown fat CD47 in age-related metabolic homeostasis, inducible brown adipocyte-specific cd47 deficient mice were generated by crossbreeding cd47 floxed mice with UCP1-CreERT2 mice and characterized in this study. Efficient knockdown of CD47 in brown fat was achieved in both male and female mice through tamoxifen administration. Intriguingly, our findings indicated that male mice lacking CD47 in brown fat displayed a notable reduction in body weight starting at 23 weeks of age when housed at a temperature of 22 °C, in comparison to control mice. This reduction in weight was accompanied by improved glucose tolerance. Remarkably, this phenotype persisted even when the male mice were housed under thermoneutral conditions (30 °C). Conversely, female knockout mice did not exhibit significant changes in weight throughout the study. In addition to the enhanced glucose homeostasis, brown fat CD47 deficiency in male mice also prevented age-related hypertriglyceridemia and non-alcoholic fatty liver disease. Furthermore, the brown fat tissue of male knockout mice exhibited reduced whitening, while maintaining comparable levels of thermogenic markers. This suggests the involvement of a thermogenesis-independent mechanism. Altogether, these findings highlight a sex difference in the impact of brown adipocyte CD47 deficiency on age-related weight changes and glucose homeostasis.
Collapse
Affiliation(s)
- Dong Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA; Lexington VA Medical Center, Lexington, KY, 40502, USA
| | - Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA; Lexington VA Medical Center, Lexington, KY, 40502, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA; Lexington VA Medical Center, Lexington, KY, 40502, USA.
| |
Collapse
|
8
|
Wu W, Liu K, You Z, Zhang J. MiR-196b-3p and miR-450b-3p are key regulators of adipogenesis in porcine intramuscular and subcutaneous adipocytes. BMC Genomics 2023; 24:360. [PMID: 37369998 DOI: 10.1186/s12864-023-09477-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND As components of white adipose tissue, porcine intramuscular (IM) and subcutaneous (SC) adipocytes undergo similar differentiation and adipogenesis processes. However, the adipogenic capacity of IM adipocytes is weaker than that of SC adipocytes. Identifying key regulators underlying this difference between IM and SC adipocytes will benefit pig breeding. RESULTS In this study, we used BGISEQ-500 sequencing technology to analyze the expression of small RNAs in primary cultured IM and SC adipocytes on day 8 after adipogenic induction, and found 32-fold higher miR-196b-3p expression, as well as 8-fold lower miR-450b-3p expression in IM adipocytes than in SC adipocytes. Functional studies revealed that miR-196b-3p inhibits adipogenesis by targeting CD47 via the AMPK signaling pathway, and its effect was attenuated by the specific p-AMPKα activator AICAR. We also found that miR-450b-3p promotes adipogenesis by targeting SIRT1 via the Wnt/β-catenin signaling pathway, and its effect was weakened by the Wnt/β-catenin signaling activator LiCl. CONCLUSIONS Our findings suggest that miR-196b-3p and miR-450b-3p are novel key regulatory factors that play opposite roles in porcine adipogenesis, helping us decipher the regulatory differences between porcine IM and SC fat deposition.
Collapse
Affiliation(s)
- Wenjing Wu
- College of Biological and Chemical Engineering, Jiaxing University, Jiaxing Zhejiang, 314000, China
| | - Keke Liu
- College of Biological and Chemical Engineering, Jiaxing University, Jiaxing Zhejiang, 314000, China
- College of Agronomy and Biotechnology, Hebei Normal University of Science and Technology, Qin Huangdao Hebei, 066000, China
| | - Zhongyu You
- College of Biological and Chemical Engineering, Jiaxing University, Jiaxing Zhejiang, 314000, China
| | - Jin Zhang
- College of Biological and Chemical Engineering, Jiaxing University, Jiaxing Zhejiang, 314000, China.
| |
Collapse
|
9
|
Li JH, Hepworth MR, O'Sullivan TE. Regulation of systemic metabolism by tissue-resident immune cell circuits. Immunity 2023; 56:1168-1186. [PMID: 37315533 PMCID: PMC10321269 DOI: 10.1016/j.immuni.2023.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023]
Abstract
Recent studies have demonstrated that tissue homeostasis and metabolic function are dependent on distinct tissue-resident immune cells that form functional cell circuits with structural cells. Within these cell circuits, immune cells integrate cues from dietary contents and commensal microbes in addition to endocrine and neuronal signals present in the tissue microenvironment to regulate structural cell metabolism. These tissue-resident immune circuits can become dysregulated during inflammation and dietary overnutrition, contributing to metabolic diseases. Here, we review the evidence describing key cellular networks within and between the liver, gastrointestinal tract, and adipose tissue that control systemic metabolism and how these cell circuits become dysregulated during certain metabolic diseases. We also identify open questions in the field that have the potential to enhance our understanding of metabolic health and disease.
Collapse
Affiliation(s)
- Joey H Li
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA; Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
10
|
Erdem N, Chen KT, Qi M, Zhao Y, Wu X, Garcia I, Ku HT, Montero E, Al-Abdullah IH, Kandeel F, Roep BO, Isenberg JS. Thrombospondin-1, CD47, and SIRPα display cell-specific molecular signatures in human islets and pancreata. Am J Physiol Endocrinol Metab 2023; 324:E347-E357. [PMID: 36791324 PMCID: PMC11967708 DOI: 10.1152/ajpendo.00221.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/10/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
Thrombospondin-1 (TSP1) is a secreted protein minimally expressed in health but increased in disease and age. TSP1 binds to the cell membrane receptor CD47, which itself engages signal regulatory protein α (SIRPα), and the latter creates a checkpoint for immune activation. Individuals with cancer administered checkpoint-blocking molecules developed insulin-dependent diabetes. Relevant to this, CD47 blocking antibodies and SIRPα fusion proteins are in clinical trials. We characterized the molecular signature of TSP1, CD47, and SIRPα in human islets and pancreata. Fresh islets and pancreatic tissue from nondiabetic individuals were obtained. The expression of THBS1, CD47, and SIRPA was determined using single-cell mRNA sequencing, immunofluorescence microscopy, Western blot, and flow cytometry. Islets were exposed to diabetes-affiliated inflammatory cytokines and changes in protein expression were determined. CD47 mRNA was expressed in all islet cell types. THBS1 mRNA was restricted primarily to endothelial and mesenchymal cells, whereas SIRPA mRNA was found mostly in macrophages. Immunofluorescence staining showed CD47 protein expressed by β cells and present in the exocrine pancreas. TSP1 and SIRPα proteins were not seen in islets or the exocrine pancreas. Western blot and flow cytometry confirmed immunofluorescent expression patterns. Importantly, human islets produced substantial quantities of secreted TSP1. Human pancreatic exocrine and endocrine tissue expressed CD47, whereas fresh islets displayed cell surface CD47 and secreted TSP1 at baseline and in inflammation. These findings suggest unexpected effects on islets from agents that intersect TSP1-CD47-SIRPα.NEW & NOTEWORTHY CD47 is a cell surface receptor with two primary ligands, soluble thrombospondin-1 (TSP1) and cell surface signal regulatory protein alpha (SIRPα). Both interactions provide checkpoints for immune cell activity. We determined that fresh human islets display CD47 and secrete TSP1. However, human islet endocrine cells lack SIRPα. These gene signatures are likely important given the increasing use of CD47 and SIRPα blocking molecules in individuals with cancer.
Collapse
Affiliation(s)
- Neslihan Erdem
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
- Department of Molecular & Cellular Endocrinology, City of Hope National Medical Center, Duarte, California, United States
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Kuan-Tsen Chen
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Meirigeng Qi
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Yuqi Zhao
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Isaac Garcia
- Department of Molecular & Cellular Endocrinology, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Hsun Teresa Ku
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Enrique Montero
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Ismail H Al-Abdullah
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Bart O Roep
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| | - Jeffrey S Isenberg
- Department of Diabetes Complications & Metabolism, City of Hope National Medical Center, Duarte, California, United States
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, California, United States
| |
Collapse
|
11
|
Zhao W, Shen B, Cheng Q, Zhou Y, Chen K. Roles of TSP1-CD47 signaling pathway in senescence of endothelial cells: cell cycle, inflammation and metabolism. Mol Biol Rep 2023; 50:4579-4585. [PMID: 36897523 DOI: 10.1007/s11033-023-08357-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023]
Abstract
Endothelial cells (ECs) serve as a barrier with forming a monolayer lining in the surface of vascular system. Many mature cell types are post-mitotic like neurons, but ECs have the ability to grow during angiogenesis. Vascular endothelial growth factor (VEGF) stimulates growth of vascular ECs derived from arteries, veins, and lymphatics and induces angiogenesis. Senescence of ECs is regarded as a key contributor in aging-induced vascular dysfunction via evoking increase of ECs permeability, impairment of angiogenesis and vascular repair. Several genomics and proteomics studies on ECs senescence reported changes in gene and protein expression that directly correlate with vascular systemic disorder. CD47 functions as a signaling receptor for secreted matricellular protein thrombospondin-1 (TSP1) and plays an important role in several fundamental cellular functions, including proliferation, apoptosis, inflammation, and atherosclerotic response. TSP1-CD47 signaling is upregulated with age in ECs, concurrent with suppression of key self-renewal genes. Recent studies indicate that CD47 is involved in regulation of senescence, self-renewal and inflammation. In this review, we highlight the functions of CD47 in senescent ECs, including modulation of cell cycle, mediation of inflammation and metabolism by the experimental studies, which may provide CD47 as a potential therapeutic target for aging-associated vascular dysfunction.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Botao Shen
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Quanli Cheng
- Department of Cardiology, The First Hospital of Jilin University, Changchun, China
| | - Yangyang Zhou
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.
| | - Kexin Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
12
|
The Important Role of m6A-Modified circRNAs in the Differentiation of Intramuscular Adipocytes in Goats Based on MeRIP Sequencing Analysis. Int J Mol Sci 2023; 24:ijms24054817. [PMID: 36902246 PMCID: PMC10003525 DOI: 10.3390/ijms24054817] [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: 12/22/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Intramuscular fat contributes to the improvement of goat meat quality. N6-Methyladenosine (m6A)-modified circular RNAs play important roles in adipocyte differentiation and metabolism. However, the mechanisms by which m6A modifies circRNA before and after differentiation of goat intramuscular adipocytes remain poorly understood. Here, we performed methylated RNA immunoprecipitation sequencing (MeRIP-seq) and circRNA sequencing (circRNA-seq) to determine the distinctions in m6A-methylated circRNAs during goat adipocyte differentiation. The profile of m6A-circRNA showed a total of 427 m6A peaks within 403 circRNAs in the intramuscular preadipocytes group, and 428 peaks within 401 circRNAs in the mature adipocytes group. Compared with the intramuscular preadipocytes group, 75 peaks within 75 circRNAs were significantly different in the mature adipocytes group. Furthermore, the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of intramuscular preadipocytes and mature adipocytes showed that the differentially m6A-modified circRNAs were enriched in the PKG signaling pathway, endocrine and other factor-regulated calcium reabsorption, lysine degradation, etc. m6A-circRNA-miRNA-mRNA interaction networks predicted the potential m6A-circRNA regulation mechanism in different goat adipocytes. Our results indicate that there is a complicated regulatory relationship between the 12 upregulated and 7 downregulated m6A-circRNAs through 14 and 11 miRNA mediated pathways, respectively. In addition, co-analysis revealed a positive association between m6A abundance and levels of circRNA expression, such as expression levels of circRNA_0873 and circRNA_1161, which showed that m6A may play a vital role in modulating circRNA expression during goat adipocyte differentiation. These results would provide novel information for elucidating the biological functions and regulatory characteristics of m6A-circRNAs in intramuscular adipocyte differentiation and could be helpful for further molecular breeding to improve meat quality in goats.
Collapse
|
13
|
Gwag T, Li D, Ma E, Guo Z, Liang Y, Wang S. CD47 antisense oligonucleotide treatment attenuates obesity and its-associated metabolic dysfunction. Sci Rep 2023; 13:2748. [PMID: 36797364 PMCID: PMC9935863 DOI: 10.1038/s41598-023-30006-2] [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: 12/15/2022] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Previous study from our lab has revealed a new role of CD47 in regulating adipose tissue function, energy homeostasis and the development of obesity and metabolic disease in CD47 deficient mice. In this study, the therapeutic potential of an antisense oligonucleotide (ASO) targeting to CD47 in obesity and its-associated complications was determined in two obese mouse models (diet induced and genetic models). In diet induced obesity, male C57BL6 mice were fed with high fat (HF) diet to induce obesity and then treated with CD47ASO or control ASO for 8 weeks. In genetic obese mouse model, male six-week old ob/ob mice were treated with ASOs for 9 weeks. We found that CD47ASO treatment reduced HF diet-induced weight gain, decreased fat mass, prevented dyslipidemia, and improved glucose tolerance. These changes were accompanied by reduced inflammation in white adipose tissue and decreased hepatic steatosis. This protection was also seen in CD47ASO treated ob/ob mice. Mechanistically, CD47ASO treatment increased mice physical activity and energy expenditure, contributing to weight loss and improved metabolic outcomes in obese mice. Collectively, these findings suggest that CD47ASO might serve as a new treatment option for obesity and its-associated metabolic complications.
Collapse
Affiliation(s)
- Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Wethington Bldg. Room 583, 900 S. Limestone Street, Lexington, KY, 40536, USA
- Lexington Veterans Affairs Medical Center, Lexington, KY, 40502, USA
| | - Dong Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Wethington Bldg. Room 583, 900 S. Limestone Street, Lexington, KY, 40536, USA
- Lexington Veterans Affairs Medical Center, Lexington, KY, 40502, USA
| | - Eric Ma
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Wethington Bldg. Room 583, 900 S. Limestone Street, Lexington, KY, 40536, USA
| | - Zhenheng Guo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Wethington Bldg. Room 583, 900 S. Limestone Street, Lexington, KY, 40536, USA
| | - Ying Liang
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Wethington Bldg. Room 583, 900 S. Limestone Street, Lexington, KY, 40536, USA.
- Lexington Veterans Affairs Medical Center, Lexington, KY, 40502, USA.
| |
Collapse
|
14
|
Gao L, He Z, Wu Y. Advances in Anti-metabolic Disease Treatments Targeting CD47. Curr Pharm Des 2022; 28:3720-3728. [PMID: 36201266 DOI: 10.2174/1381612828666221006123144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 01/28/2023]
Abstract
Metabolic disorders include a cluster of conditions that result from hyperglycemia, hyperlipidemia, insulin resistance, obesity, and hepatic steatosis, which cause the dysfunction of immune cells and innate cells, such as macrophages, natural killer cells, vascular endothelial cells, hepatocytes, and human kidney tubular epithelial cells. Besides targeting the derangements in lipid metabolism, therapeutic modulations to regulate abnormal responses in the immune system and innate cell dysfunctions may prove to be promising strategies in the management of metabolic diseases. In recent years, several targets have been explored for the CD47 molecule (CD47), a glycosylated protein, which was originally reported to transmit an anti-phagocytic signal known as "don't eat me" in the atherosclerotic environment, hindering the efferocytosis of immune cells and promoting arterial plaque accumulation. Subsequently, the role of CD47 has been explored in obesity, fatty liver, and lipotoxic nephropathy, and its utility as a therapeutic target has been investigated using anti-CD47 antibodies or inhibitors of the THBS1/CD47 axis and the CD47/SIRPα signaling pathway. This review summarizes the mechanisms of action of CD47 in different cell types during metabolic diseases and the clinical research progress to date, providing a reference for the comprehensive targeting of CD47 to treat metabolic diseases and the devising of potential improvements to possible side effects.
Collapse
Affiliation(s)
- Li Gao
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.,Center for Scientific Research of Anhui Medical University, Hefei 230022, China
| | - Zhe He
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yonggui Wu
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.,Center for Scientific Research of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
15
|
Shi H, Wang X, Li F, Gerlach BD, Yurdagul A, Moore MP, Zeldin S, Zhang H, Cai B, Zheng Z, Valenti L, Tabas I. CD47-SIRPα axis blockade in NASH promotes necroptotic hepatocyte clearance by liver macrophages and decreases hepatic fibrosis. Sci Transl Med 2022; 14:eabp8309. [PMID: 36417485 PMCID: PMC10199725 DOI: 10.1126/scitranslmed.abp8309] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Necroptosis contributes to hepatocyte death in nonalcoholic steatohepatitis (NASH), but the fate and roles of necroptotic hepatocytes (necHCs) in NASH remain unknown. We show here that the accumulation of necHCs in human and mouse NASH liver is associated with an up-regulation of the "don't-eat-me" ligand CD47 on necHCs, but not on apoptotic hepatocytes, and an increase in the CD47 receptor SIRPα on liver macrophages, consistent with impaired macrophage-mediated clearance of necHCs. In vitro, necHC clearance by primary liver macrophages was enhanced by treatment with either anti-CD47 or anti-SIRPα. In a proof-of-concept mouse model of inducible hepatocyte necroptosis, anti-CD47 antibody treatment increased necHC uptake by liver macrophages and inhibited markers of hepatic stellate cell (HSC) activation, which is responsible for liver fibrogenesis. Treatment of two mouse models of diet-induced NASH with anti-CD47, anti-SIRPα, or AAV8-H1-shCD47 to silence CD47 in hepatocytes increased the uptake of necHC by liver macrophages and decreased markers of HSC activation and liver fibrosis. Anti-SIRPα treatment avoided the adverse effect of anemia found in anti-CD47-treated mice. These findings provide evidence that impaired clearance of necHCs by liver macrophages due to CD47-SIRPα up-regulation contributes to fibrotic NASH, and suggest therapeutic blockade of the CD47-SIRPα axis as a strategy to decrease the accumulation of necHCs in NASH liver and dampen the progression of hepatic fibrosis.
Collapse
Affiliation(s)
- Hongxue Shi
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Fang Li
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brennan D. Gerlach
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mary P. Moore
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sharon Zeldin
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hanrui Zhang
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Bishuang Cai
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ze Zheng
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano and Fondazione Ca’ Granda Ospedale Maggiore Policlinico Milano, Milano 20122, Italy
| | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
16
|
Krieg L, Didt K, Karkossa I, Bernhart SH, Kehr S, Subramanian N, Lindhorst A, Schaudinn A, Tabei S, Keller M, Stumvoll M, Dietrich A, von Bergen M, Stadler PF, Laurencikiene J, Krüger M, Blüher M, Gericke M, Schubert K, Kovacs P, Chakaroun R, Massier L. Multiomics reveal unique signatures of human epiploic adipose tissue related to systemic insulin resistance. Gut 2022; 71:2179-2193. [PMID: 34598978 PMCID: PMC9554031 DOI: 10.1136/gutjnl-2021-324603] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Human white adipose tissue (AT) is a metabolically active organ with distinct depot-specific functions. Despite their locations close to the gastrointestinal tract, mesenteric AT and epiploic AT (epiAT) have only scarcely been investigated. Here, we aim to characterise these ATs in-depth and estimate their contribution to alterations in whole-body metabolism. DESIGN Mesenteric, epiploic, omental and abdominal subcutaneous ATs were collected from 70 patients with obesity undergoing Roux-en-Y gastric bypass surgery. The metabolically well-characterised cohort included nine subjects with insulin sensitive (IS) obesity, whose AT samples were analysed in a multiomics approach, including methylome, transcriptome and proteome along with samples from subjects with insulin resistance (IR) matched for age, sex and body mass index (n=9). Findings implying differences between AT depots in these subgroups were validated in the entire cohort (n=70) by quantitative real-time PCR. RESULTS While mesenteric AT exhibited signatures similar to those found in the omental depot, epiAT was distinct from all other studied fat depots. Multiomics allowed clear discrimination between the IS and IR states in all tissues. The highest discriminatory power between IS and IR was seen in epiAT, where profound differences in the regulation of developmental, metabolic and inflammatory pathways were observed. Gene expression levels of key molecules involved in AT function, metabolic homeostasis and inflammation revealed significant depot-specific differences with epiAT showing the highest expression levels. CONCLUSION Multi-omics epiAT signatures reflect systemic IR and obesity subphenotypes distinct from other fat depots. Our data suggest a previously unrecognised role of human epiploic fat in the context of obesity, impaired insulin sensitivity and related diseases.
Collapse
Affiliation(s)
- Laura Krieg
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Konrad Didt
- Department for Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Leipzig, Germany
| | - Isabel Karkossa
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Stephan H Bernhart
- Faculty of Mathematics and Computer Science, Department of Computer Science and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Stephanie Kehr
- Faculty of Mathematics and Computer Science, Department of Computer Science and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
| | | | - Andreas Lindhorst
- Faculty of Medicine, Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Alexander Schaudinn
- Department of Diagnostic and Interventional Radiology, University Hospital Leipzig, Leipzig, Germany
| | - Shirin Tabei
- Institute of Endocrinology and Diabetes, University of Lübeck, Lübeck, Germany
| | - Maria Keller
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III – Endocrinology, Nephrology and Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Arne Dietrich
- Clinic for Visceral, Transplantation and Thorax and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany,Faculty of Life Science, Institute of Biochemistry, University of Leipzig, Leipzig, Germany
| | - Peter F Stadler
- Faculty of Mathematics and Computer Science, Department of Computer Science and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany,Max Planck Institute for Mathematics in the Sciences, Leipzig, Germany
| | | | - Martin Krüger
- Faculty of Medicine, Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, Leipzig, Germany,Medical Department III – Endocrinology, Nephrology and Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Martin Gericke
- Faculty of Medicine, Institute of Anatomy, University of Leipzig, Leipzig, Germany,Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Kristin Schubert
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research (UFZ), Leipzig, Germany
| | - Peter Kovacs
- Medical Department III – Endocrinology, Nephrology and Rheumatology, University of Leipzig Medical Center, Leipzig, Germany,Deutsches Zentrum für Diabetesforschung eV, Neuherberg, Germany
| | - Rima Chakaroun
- Medical Department III - Endocrinology, Nephrology and Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Lucas Massier
- Department of Medicine (H7), Karolinska Institutet, Stockholm, Sweden .,Medical Department III - Endocrinology, Nephrology and Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
17
|
Bronson SM, Westwood B, Cook KL, Emenaker NJ, Chappell MC, Roberts DD, Soto-Pantoja DR. Discrete Correlation Summation Clustering Reveals Differential Regulation of Liver Metabolism by Thrombospondin-1 in Low-Fat and High-Fat Diet-Fed Mice. Metabolites 2022; 12:1036. [PMID: 36355119 PMCID: PMC9697255 DOI: 10.3390/metabo12111036] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 08/08/2023] Open
Abstract
Thrombospondin-1 (TSP1) is a matricellular protein with many important roles in mediating carcinogenesis, fibrosis, leukocyte recruitment, and metabolism. We have previously shown a role of diet in the absence of TSP1 in liver metabolism in the context of a colorectal cancer model. However, the metabolic implications of TSP1 regulation by diet in the liver metabolism are currently understudied. Therefore Discrete correlation summation (DCS) was used to re-interrogate data and determine the metabolic alterations of TSP1 deficiency in the liver, providing new insights into the role of TSP1 in liver injury and the progression of liver pathologies such as nonalcoholic fatty liver disease (NAFLD). DCS analysis provides a straightforward approach to rank covariance and data clustering when analyzing complex data sets. Using this approach, our previous liver metabolite data was re-analyzed by comparing wild-type (WT) and Thrombospondin-1 null (Thbs1-/-) mice, identifying changes driven by genotype and diet. Principal component analysis showed clustering of animals by genotype regardless of diet, indicating that TSP1 deficiency alters metabolite handling in the liver. High-fat diet consumption significantly altered over 150 metabolites in the Thbs1-/- livers versus approximately 90 in the wild-type livers, most involved in amino acid metabolism. The absence of Thbs1 differentially regulated tryptophan and tricarboxylic acid cycle metabolites implicated in the progression of NAFLD. Overall, the lack of Thbs1 caused a significant shift in liver metabolism with potential implications for liver injury and the progression of NAFLD.
Collapse
Affiliation(s)
- Steven M. Bronson
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Section of Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Brian Westwood
- Department of Surgery, Hypertension & Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Katherine L. Cook
- Department of Surgery, Hypertension & Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27101, USA
| | - Nancy J. Emenaker
- Nutritional Science Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark C. Chappell
- Department of Surgery, Hypertension & Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - David D. Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David R. Soto-Pantoja
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Surgery, Hypertension & Vascular Research Center, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27101, USA
| |
Collapse
|
18
|
Delannoy-Bruno O, Desai C, Castillo JJ, Couture G, Barve RA, Lombard V, Henrissat B, Cheng J, Han N, Hayashi DK, Meynier A, Vinoy S, Lebrilla CB, Marion S, Heath AC, Barratt MJ, Gordon JI. An approach for evaluating the effects of dietary fiber polysaccharides on the human gut microbiome and plasma proteome. Proc Natl Acad Sci U S A 2022; 119:e2123411119. [PMID: 35533274 PMCID: PMC9171781 DOI: 10.1073/pnas.2123411119] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
Increases in snack consumption associated with Westernized lifestyles provide an opportunity to introduce nutritious foods into poor diets. We describe two 10-wk-long open label, single group assignment human studies that measured the effects of two snack prototypes containing fiber preparations from two sustainable and scalable sources; the byproducts remaining after isolation of protein from the endosperm of peas and the vesicular pulp remaining after processing oranges for the manufacture of juices. The normal diets of study participants were supplemented with either a pea- or orange fiber-containing snack. We focused our analysis on quantifying the abundances of genes encoding carbohydrate-active enzymes (CAZymes) (glycoside hydrolases and polysaccharide lyases) in the fecal microbiome, mass spectrometric measurements of glycan structures (glycosidic linkages) in feces, plus aptamer-based assessment of levels of 1,300 plasma proteins reflecting a broad range of physiological functions. Computational methods for feature selection identified treatment-discriminatory changes in CAZyme genes that correlated with alterations in levels of fiber-associated glycosidic linkages; these changes in turn correlated with levels of plasma proteins representing diverse biological functions, including transforming growth factor type β/bone morphogenetic protein-mediated fibrosis, vascular endothelial growth factor-related angiogenesis, P38/MAPK-associated immune cell signaling, and obesity-associated hormonal regulators. The approach used represents a way to connect changes in consumer microbiomes produced by specific fiber types with host responses in the context of varying background diets.
Collapse
Affiliation(s)
- Omar Delannoy-Bruno
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110
| | - Chandani Desai
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110
| | - Juan J. Castillo
- Department of Chemistry, University of California, Davis, CA 95616
| | - Garret Couture
- Department of Chemistry, University of California, Davis, CA 95616
| | - Ruteja A. Barve
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Vincent Lombard
- Architecture et Fonction des Macromolécules Biologiques, Centre National de la Recherche Scientifique and Aix-Marseille Université, 13288 Marseille cedex 9, France
| | - Bernard Henrissat
- Department of Biotechnology and Biomedicine (DTU Bioengineering), Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jiye Cheng
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110
| | - Nathan Han
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110
| | | | | | | | | | - Stacey Marion
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Andrew C. Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J. Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110
| | - Jeffrey I. Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
19
|
Gwag T, Ma E, Zhou C, Wang S. Anti-CD47 antibody treatment attenuates liver inflammation and fibrosis in experimental non-alcoholic steatohepatitis models. Liver Int 2022; 42:829-841. [PMID: 35129307 PMCID: PMC9101015 DOI: 10.1111/liv.15182] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 01/07/2022] [Accepted: 02/01/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND & AIMS With the epidemic burden of obesity and metabolic diseases, nonalcoholic fatty liver disease (NAFLD) including steatohepatitis (NASH) has become the most common chronic liver disease in the western world. NASH may progress to cirrhosis and hepatocellular carcinoma. Currently, no treatment is available for NASH. Therefore, finding a therapy for NAFLD/NASH is in urgent need. Previously we have demonstrated that mice lacking CD47 or its ligand thrombospondin1 (TSP1) are protected from obesity-associated NALFD. This suggests that CD47 blockade might be a novel treatment for obesity-associated metabolic disease. Thus, in this study, the therapeutic potential of an anti-CD47 antibody in NAFLD progression was determined. METHODS Both diet-induced NASH mouse model and human NASH organoid model were utilized in this study. NASH was induced in mice by feeding with diet enriched with fat, fructose and cholesterol (AMLN diet) for 20 weeks and then treated with anti-CD47 antibody or control IgG for 4 weeks. Body weight, body composition and liver phenotype were analysed. RESULTS We found that anti-CD47 antibody treatment did not affect mice body weight, fat mass or liver steatosis. However, liver immune cell infiltration, inflammation and fibrosis were significantly reduced by anti-CD47 antibody treatment. In vitro data further showed that CD47 blockade prevented hepatic stellate cell activation and NASH progression in a human NASH organoid model. CONCLUSION Collectively, these data suggest that anti-CD47 antibody might be a new therapeutic option for obesity-associated NASH and liver fibrosis.
Collapse
Affiliation(s)
- Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, and Lexington VA Medical Center, Lexington KY 40502
| | - Eric Ma
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, and Lexington VA Medical Center, Lexington KY 40502
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, and Lexington VA Medical Center, Lexington KY 40502.,To whom correspondence should be addressed: Shuxia Wang, MD, PhD, Department of Pharmacology and Nutritional Sciences, University of Kentucky, Wethington Bldg. Room 583, 900 S. Limestone Street, Lexington, KY 40536. Tel: 859-218-1367, Fax: 859-257-3646,
| |
Collapse
|
20
|
Li D, Gwag T, Wang S. Absence of CD47 maintains brown fat thermogenic capacity and protects mice from aging-related obesity and metabolic disorder. Biochem Biophys Res Commun 2021; 575:14-19. [PMID: 34454175 DOI: 10.1016/j.bbrc.2021.08.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/23/2021] [Indexed: 02/04/2023]
Abstract
Brown and beige adipocytes burn energy to produce heat and could serve as a therapeutic target to counteract metabolic diseases including obesity and type 2 diabetes. Aging is associated with reduced brown fat mass and thermogenic capacity and a risk factor for metabolic diseases. Our previous studies implicated a role for CD47 in regulating brown fat function and energy balance in young adult animals. In this study, we further determined its role in natural aging related metabolic disorders. The results demonstrated that aged CD47 deficient mice (under normal chow diet) had reduced body weight and fat mass, and improved glucose tolerance as compared to aged wild type (WT) mice. Indirect calorimetry result showed that food intake and total activity were comparable between two genotypes. However, CD47 deficient mice had increased energy expenditure and better cold tolerance, accompanied by increased white adipose tissue browning and well-maintained juvenile morphology of brown adipose tissue (BAT). Moreover, transcriptome (RNA-seq) and pathway enrichment analysis revealed that BAT from aged CD47 deficient mice had upregulated genes involving in mitochondria oxidative phosphorylation, thermogenesis, fatty acid metabolism, and valine, leucine and isoleucine (BCAA) degradation, indicating the activated BAT status in aged CD47 deficient mice. Collectively, these data suggest that blocking CD47 signaling protects mice from natural aging-associated obesity and glucose intolerance, partially though activation and expansion of the thermogenic machinery, further supporting that CD47 maybe a potential target for aging related metabolic disorder.
Collapse
Affiliation(s)
- Dong Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA; Lexington VA Medical Center, Lexington, KY, 40502, USA
| | - Taesik Gwag
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA; Lexington VA Medical Center, Lexington, KY, 40502, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA; Lexington VA Medical Center, Lexington, KY, 40502, USA.
| |
Collapse
|
21
|
A Potential Role of the CD47/SIRPalpha Axis in COVID-19 Pathogenesis. Curr Issues Mol Biol 2021; 43:1212-1225. [PMID: 34698067 PMCID: PMC8929144 DOI: 10.3390/cimb43030086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
The coronavirus SARS-CoV-2 is the cause of the ongoing COVID-19 pandemic. Most SARS-CoV-2 infections are mild or even asymptomatic. However, a small fraction of infected individuals develops severe, life-threatening disease, which is caused by an uncontrolled immune response resulting in hyperinflammation. However, the factors predisposing individuals to severe disease remain poorly understood. Here, we show that levels of CD47, which is known to mediate immune escape in cancer and virus-infected cells, are elevated in SARS-CoV-2-infected Caco-2 cells, Calu-3 cells, and air-liquid interface cultures of primary human bronchial epithelial cells. Moreover, SARS-CoV-2 infection increases SIRPalpha levels, the binding partner of CD47, on primary human monocytes. Systematic literature searches further indicated that known risk factors such as older age and diabetes are associated with increased CD47 levels. High CD47 levels contribute to vascular disease, vasoconstriction, and hypertension, conditions that may predispose SARS-CoV-2-infected individuals to COVID-19-related complications such as pulmonary hypertension, lung fibrosis, myocardial injury, stroke, and acute kidney injury. Hence, age-related and virus-induced CD47 expression is a candidate mechanism potentially contributing to severe COVID-19, as well as a therapeutic target, which may be addressed by antibodies and small molecules. Further research will be needed to investigate the potential involvement of CD47 and SIRPalpha in COVID-19 pathology. Our data should encourage other research groups to consider the potential relevance of the CD47/ SIRPalpha axis in their COVID-19 research.
Collapse
|
22
|
Kan K, Mu Y, Bouschbacher M, Sticht C, Kuch N, Sigl M, Rahbari N, Gretz N, Pallavi P, Keese M. Biphasic Effects of Blue Light Irradiation on Human Umbilical Vein Endothelial Cells. Biomedicines 2021; 9:biomedicines9070829. [PMID: 34356893 PMCID: PMC8301484 DOI: 10.3390/biomedicines9070829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 11/22/2022] Open
Abstract
Blue light regulates biological function in various cells, such as proliferation, oxidative stress, and cell death. We employed blue light illumination on human umbilical vein endothelial cells utilizing a LED device at 453 nm wavelength and revealed a novel biphasic response on human umbilical vein endothelial cells (HUVECs). The results showed that low fluence blue light irradiation promoted the fundamental cell activities, including cell viability, migration and angiogenesis by activating the angiogenic pathways such as the VEGF signaling pathway. In contrast, high fluence illumination caused the opposite effect on those activities by upregulating pro-apoptotic signaling cascades like ferroptosis, necroptosis and the p53 signaling pathways. Our results provide an underlying insight into photobiomodulation by blue light and may help to implement potential treatment strategies for treating angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Kejia Kan
- Department of Vascular Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (K.K.); (Y.M.); (N.K.)
- European Center of Angioscience ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Yifei Mu
- Department of Vascular Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (K.K.); (Y.M.); (N.K.)
| | | | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Natalia Kuch
- Department of Vascular Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (K.K.); (Y.M.); (N.K.)
| | - Martin Sigl
- First Department of Medicine, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Nuh Rahbari
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Norbert Gretz
- Medical Research Centre, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Prama Pallavi
- Department of Vascular Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (K.K.); (Y.M.); (N.K.)
- European Center of Angioscience ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Correspondence: (P.P.); (M.K.); Tel.: +49-621-383-4057 (P.P.); +49-621-383-1501 (M.K.)
| | - Michael Keese
- Department of Vascular Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (K.K.); (Y.M.); (N.K.)
- European Center of Angioscience ECAS, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Correspondence: (P.P.); (M.K.); Tel.: +49-621-383-4057 (P.P.); +49-621-383-1501 (M.K.)
| |
Collapse
|
23
|
Roberts DD, Isenberg JS. CD47 and thrombospondin-1 regulation of mitochondria, metabolism, and diabetes. Am J Physiol Cell Physiol 2021; 321:C201-C213. [PMID: 34106789 DOI: 10.1152/ajpcell.00175.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Thrombospondin-1 (TSP1) is the prototypical member of a family of secreted proteins that modulate cell behavior by engaging with molecules in the extracellular matrix and with receptors on the cell surface. CD47 is widely displayed on many, if not all, cell types and is a high-affinity TSP1 receptor. CD47 is a marker of self that limits innate immune cell activities, a feature recently exploited to enhance cancer immunotherapy. Another major role for CD47 in health and disease is to mediate TSP1 signaling. TSP1 acting through CD47 contributes to mitochondrial, metabolic, and endocrine dysfunction. Studies in animal models found that elevated TSP1 expression, acting in part through CD47, causes mitochondrial and metabolic dysfunction. Clinical studies established that abnormal TSP1 expression positively correlates with obesity, fatty liver disease, and diabetes. The unabated increase in these conditions worldwide and the availability of CD47 targeting drugs justify a closer look into how TSP1 and CD47 disrupt metabolic balance and the potential for therapeutic intervention.
Collapse
Affiliation(s)
- David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
24
|
Thrombospondin-1 CD47 Signalling: From Mechanisms to Medicine. Int J Mol Sci 2021; 22:ijms22084062. [PMID: 33920030 PMCID: PMC8071034 DOI: 10.3390/ijms22084062] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/19/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Recent advances provide evidence that the cellular signalling pathway comprising the ligand-receptor duo of thrombospondin-1 (TSP1) and CD47 is involved in mediating a range of diseases affecting renal, vascular, and metabolic function, as well as cancer. In several instances, research has barely progressed past pre-clinical animal models of disease and early phase 1 clinical trials, while for cancers, anti-CD47 therapy has emerged from phase 2 clinical trials in humans as a crucial adjuvant therapeutic agent. This has important implications for interventions that seek to capitalize on targeting this pathway in diseases where TSP1 and/or CD47 play a role. Despite substantial progress made in our understanding of this pathway in malignant and cardiovascular disease, knowledge and translational gaps remain regarding the role of this pathway in kidney and metabolic diseases, limiting identification of putative drug targets and development of effective treatments. This review considers recent advances reported in the field of TSP1-CD47 signalling, focusing on several aspects including enzymatic production, receptor function, interacting partners, localization of signalling, matrix-cellular and cell-to-cell cross talk. The potential impact that these newly described mechanisms have on health, with a particular focus on renal and metabolic disease, is also discussed.
Collapse
|
25
|
Norman-Burgdolf H, Li D, Sullivan P, Wang S. CD47 differentially regulates white and brown fat function. Biol Open 2020; 9:bio056747. [PMID: 33328190 PMCID: PMC7758621 DOI: 10.1242/bio.056747] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/19/2020] [Indexed: 01/18/2023] Open
Abstract
Mechanisms that enhance energy expenditure are attractive therapeutic targets for obesity. Previously we have demonstrated that mice lacking cd47 are leaner, exhibit increased energy expenditure, and are protected against diet-induced obesity. In this study, we further defined the physiological role of cd47 deficiency in regulating mitochondrial function and energy expenditure in both white and brown adipose tissue. We observed that cd47 deficient mice (under normal chow diet) had comparable amount of white fat mass but reduced white adipocyte size as compared to wild-type mice. Subsequent ex vivo and in vitro studies suggest enhanced lipolysis, and not impaired lipogenesis or energy utilization, contributes to this phenotype. In contrast to white adipose tissue, there were no obvious morphological differences in brown adipose tissue between wild-type and knockout mice. However, mitochondria isolated from brown fat of cd47 deficient mice had significantly higher rates of free fatty acid-mediated uncoupling. This suggests that enhanced fuel availability via white adipose tissue lipolysis may perpetuate elevated brown adipose tissue energy expenditure and contributes to the lean phenotype observed in cd47 deficient mice.
Collapse
Affiliation(s)
- Heather Norman-Burgdolf
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Dietetics and Human Nutrition, University of Kentucky, Lexington, KY 40536, USA
| | - Dong Li
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Research and Development, Lexington VA Medical Center, Lexington KY 40502, USA
| | - Patrick Sullivan
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Department of Research and Development, Lexington VA Medical Center, Lexington KY 40502, USA
| | - Shuxia Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
- Department of Research and Development, Lexington VA Medical Center, Lexington KY 40502, USA
| |
Collapse
|
26
|
Ghimire K, Li Y, Chiba T, Julovi SM, Li J, Ross MA, Straub AC, O’Connell PJ, Rüegg C, Pagano PJ, Isenberg JS, Rogers NM. CD47 Promotes Age-Associated Deterioration in Angiogenesis, Blood Flow and Glucose Homeostasis. Cells 2020; 9:E1695. [PMID: 32679764 PMCID: PMC7407670 DOI: 10.3390/cells9071695] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 02/06/2023] Open
Abstract
The aged population is currently at its highest level in human history and is expected to increase further in the coming years. In humans, aging is accompanied by impaired angiogenesis, diminished blood flow and altered metabolism, among others. A cellular mechanism that impinges upon these manifestations of aging can be a suitable target for therapeutic intervention. Here we identify cell surface receptor CD47 as a novel age-sensitive driver of vascular and metabolic dysfunction. With the natural aging process, CD47 and its ligand thrombospondin-1 were increased, concurrent with a reduction of self-renewal transcription factors OCT4, SOX2, KLF4 and cMYC (OSKM) in arteries from aged wild-type mice and older human subjects compared to younger controls. These perturbations were prevented in arteries from aged CD47-null mice. Arterial endothelial cells isolated from aged wild-type mice displayed cellular exhaustion with decreased proliferation, migration and tube formation compared to cells from aged CD47-null mice. CD47 suppressed ex vivo sprouting, in vivo angiogenesis and skeletal muscle blood flow in aged wild-type mice. Treatment of arteries from older humans with a CD47 blocking antibody mitigated the age-related deterioration in angiogenesis. Finally, aged CD47-null mice were resistant to age- and diet-associated weight gain, glucose intolerance and insulin desensitization. These results indicate that the CD47-mediated signaling maladapts during aging to broadly impair endothelial self-renewal, angiogenesis, perfusion and glucose homeostasis. Our findings provide a strong rationale for therapeutically targeting CD47 to minimize these dysfunctions during aging.
Collapse
Affiliation(s)
- Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Rd, Sydney 2145, NSW, Australia; (S.M.J.); (J.L.); (P.J.O.)
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; (Y.L.); (T.C.); (A.C.S.); (P.J.P.)
| | - Yao Li
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; (Y.L.); (T.C.); (A.C.S.); (P.J.P.)
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Takuto Chiba
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; (Y.L.); (T.C.); (A.C.S.); (P.J.P.)
| | - Sohel M. Julovi
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Rd, Sydney 2145, NSW, Australia; (S.M.J.); (J.L.); (P.J.O.)
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Rd, Sydney 2145, NSW, Australia; (S.M.J.); (J.L.); (P.J.O.)
| | - Mark A. Ross
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, BST, 200 Lothrop Street, Pittsburgh, PA 15261, USA;
| | - Adam C. Straub
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; (Y.L.); (T.C.); (A.C.S.); (P.J.P.)
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Philip J. O’Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Rd, Sydney 2145, NSW, Australia; (S.M.J.); (J.L.); (P.J.O.)
| | - Curzio Rüegg
- Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Chemin du Musée 18, PER 17, 1700 Fribourg, Switzerland;
| | - Patrick J. Pagano
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; (Y.L.); (T.C.); (A.C.S.); (P.J.P.)
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Jeffrey S. Isenberg
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; (Y.L.); (T.C.); (A.C.S.); (P.J.P.)
- Department of Medicine, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA
| | - Natasha M. Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, 176 Hawkesbury Rd, Sydney 2145, NSW, Australia; (S.M.J.); (J.L.); (P.J.O.)
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST Starzl Tower, 200 Lothrop Street, Pittsburgh, PA 15261, USA; (Y.L.); (T.C.); (A.C.S.); (P.J.P.)
| |
Collapse
|
27
|
Feng L, Jian R, Wang Y, Yao Y, Gao M, Du H. A close relationship between Helicobacter pylori infection and the serum level of CD47 in adults. Scand J Gastroenterol 2020; 55:640-645. [PMID: 32493095 DOI: 10.1080/00365521.2020.1772863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 02/04/2023]
Abstract
Objectives: Although Helicobacter pylori (H. pylori) infection is one of the most important risk factors for gastric cancer, the molecular mechanisms underlying the progression of H. pylori-induced gastric cancer remain unclear. Previous studies have demonstrated that Integrin-associated protein (CD47) plays an important role in the development of gastric cancer. Hence, the aim of this study was to preliminarily explore the relationship between CD47 and H. pylori infection.Methods: A total of 417 adults who underwent health checkups at a hospital were recruited in 2018. Serum levels of CD47 and tumor necrosis factor-α (TNF-α) were determined using an enzyme-linked immunosorbent assay. 13C urea breath test (13C-UBT) was carried out to diagnose H. pylori infection in all participants.Results: Compared with H. pylori-negative participants, H. pylori-positive participants have higher levels of serum CD47 and TNF-α. H. pylori infection, the levels of serum TNF-α and low density lipoprotein (LDL) are the independent predictors of serum level of CD47 in adults. In addition, a potential diagnostic value of serum CD47 level for H. pylori infection has been demonstrated in our study.Conclusion: H. pylori infection is closely associated with the serum level of CD47 in adults, suggesting that H. pylori may promote gastric cancer progression by activating CD47-mediated oncogenic pathways.
Collapse
Affiliation(s)
- Lei Feng
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Rong Jian
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Yan Wang
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Yong Yao
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Mingsheng Gao
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Heng Du
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| |
Collapse
|
28
|
Isenberg JS, Roberts DD. Thrombospondin-1 in maladaptive aging responses: a concept whose time has come. Am J Physiol Cell Physiol 2020; 319:C45-C63. [PMID: 32374675 DOI: 10.1152/ajpcell.00089.2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous age-dependent alterations at the molecular, cellular, tissue and organ systems levels underlie the pathophysiology of aging. Herein, the focus is upon the secreted protein thrombospondin-1 (TSP1) as a promoter of aging and age-related diseases. TSP1 has several physiological functions in youth, including promoting neural synapse formation, mediating responses to ischemic and genotoxic stress, minimizing hemorrhage, limiting angiogenesis, and supporting wound healing. These acute functions of TSP1 generally require only transient expression of the protein. However, accumulating basic and clinical data reinforce the view that chronic diseases of aging are associated with accumulation of TSP1 in the extracellular matrix, which is a significant maladaptive contributor to the aging process. Identification of the relevant cell types that chronically produce and respond to TSP1 and the molecular mechanisms that mediate the resulting maladaptive responses could direct the development of therapeutic agents to delay or revert age-associated maladies.
Collapse
Affiliation(s)
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
29
|
Hu T, Liu H, Liang Z, Wang F, Zhou C, Zheng X, Zhang Y, Song Y, Hu J, He X, Xiao J, King RJ, Wu X, Lan P. Tumor-intrinsic CD47 signal regulates glycolysis and promotes colorectal cancer cell growth and metastasis. Am J Cancer Res 2020; 10:4056-4072. [PMID: 32226539 PMCID: PMC7086360 DOI: 10.7150/thno.40860] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/09/2020] [Indexed: 12/17/2022] Open
Abstract
Rationale: CD47 plays a vital role in the immune escape of tumor cells, but its role in regulating immune-unrelated biological processes such as proliferation and metastasis remains unclear. We seek to explore the immune-independent functions of CD47 in colorectal cancer (CRC). Methods: The expression of CD47 in CRC was determined by immunohistochemistry. The biological effect of CD47 signaling on tumor cell proliferation and metastasis was evaluated in vitro and in vivo. RNA sequencing analysis was performed to identify pivotal signaling pathways modulated by CD47. The interaction between CD47 and ENO1 was verified by co-immunoprecipitation (co-IP). The effect of CD47 on glycolytic metabolites was analyzed by seahorse XF and targeted metabolomics. Results: The expression of CD47 was upregulated and correlated to poor prognosis in CRC patients. Functional assays revealed that CD47 promoted CRC cell growth and metastasis in vitro and in vivo. Our mechanistic investigations demonstrated that CD47 interacted with ENO1 and protected it from ubiquitin-mediated degradation, subsequently promoting glycolytic activity and phosphorylation of ERK in CRC cells. Inhibition of ENO1 diminished CD47-mediated cell growth and migration. Clinically, the combined expression of CD47 and ENO1 provided reliable predictive biomarkers for the prognosis of CRC patients. Conclusions: CD47 is overexpressed in CRC, and its expression is associated with poor prognosis. Through stabilizing ENO1, CD47 enhances the aerobic glycolysis and ERK activity in CRC cells, thereby promoting the progression of CRC. Our studies reveal an unconventional role of CD47, suggesting that targeting the CD47-ENO1 axis may provide a novel therapeutic avenue for CRC.
Collapse
|
30
|
Tao HC, Chen KX, Wang X, Chen B, Zhao WO, Zheng Y, Yang YG. CD47 Deficiency in Mice Exacerbates Chronic Fatty Diet-Induced Steatohepatitis Through Its Role in Regulating Hepatic Inflammation and Lipid Metabolism. Front Immunol 2020; 11:148. [PMID: 32158445 PMCID: PMC7052326 DOI: 10.3389/fimmu.2020.00148] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammation is one of the hallmarks of non-alcoholic steatohepatitis. CD47 is a widely expressed transmembrane protein that signals through inhibitory receptor signal regulatory protein α (SIRPα) to inhibit macrophage activation and phagocytosis. In this study, we sought to investigate the role of CD47 in hepatosteatosis and fibrosis induced by a chronic high-fat diet (HFD), by comparing disease development in wild-type (WT) and CD47KO mice fed HFD for 40 weeks. The HFD induced remarkably more severe hepatic steatosis and fibrosis but less body weight gain and less subcutaneous fat accumulation in CD47KO mice compared to WT mice. Liver tissues from HFD-fed CD47KO mice exhibited enhanced inflammation characterized by increased proinflammatory cytokine production and increased nuclear factor-κB (NF-κB) activation compared to similarly fed WT mice. Although higher expression of apolipoproteins was observed in CD47KO mice compared to WT mice under a low-fat diet (LFD), HFD-fed WT and CD47KO mice showed comparably prominent downregulation of these apolipoprotein genes, suggesting that the marked difference observed in lipid accumulation and hepatosteatosis between these mice cannot be explained by changes in apolipoproteins. Like apolipoproteins, sirtuin 1 (SIRT1) and peroxisome proliferator activated receptor alpha (PPARα), which are involved in regulation of both lipid metabolism and inflammation, were more highly expressed in CD47KO than WT mice under LFD but more severely suppressed in CD47KO than in WT mice under HFD. Taken together, our results indicate that CD47 plays a significant role in the pathogenesis of HFD-induced hepatosteatosis and fibrosis through its role in regulation of inflammation and lipid metabolism.
Collapse
Affiliation(s)
- Hui-Chao Tao
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.,Cardiovascular Center, The First Hospital, Jilin University, Changchun, China
| | - Ke-Xin Chen
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.,Cardiovascular Center, The First Hospital, Jilin University, Changchun, China
| | - Xue Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Bo Chen
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Wai-Ou Zhao
- Cardiovascular Center, The First Hospital, Jilin University, Changchun, China
| | - Yang Zheng
- Cardiovascular Center, The First Hospital, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China.,National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.,Columbia Center for Translational Immunology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
31
|
Stirling ER, Cook KL, Roberts DD, Soto-Pantoja DR. Metabolomic Analysis Reveals Unique Biochemical Signatures Associated with Protection from Radiation Induced Lung Injury by Lack of cd47 Receptor Gene Expression. Metabolites 2019; 9:E218. [PMID: 31597291 PMCID: PMC6835245 DOI: 10.3390/metabo9100218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 01/10/2023] Open
Abstract
The goal of this study was to interrogate biochemical profiles manifested in mouse lung tissue originating from wild type (WT) and cd47 null mice with the aim of revealing the in vivo role of CD47 in the metabolic response to ionizing radiation, especially changes related to the known association of CD47 deficiency with increased tissue viability and survival. For this objective, we performed global metabolomic analysis in mouse lung tissue collected from (C57Bl/6 background) WT and cd47 null mice with and without exposure to 7.6 Gy whole body radiation. Principal component analysis and hierarchical clustering revealed a consistent separation between genotypes following radiation exposure. Random forest analysis also revealed a unique biochemical signature in WT and cd47 null mice following treatment. Our data show that cd47 null irradiated lung tissue activates a unique set of metabolic pathways that facilitate the handling of reactive oxygen species, lipid metabolism, nucleotide metabolism and nutrient metabolites which may be regulated by microbial processing. Given that cd47 has pleiotropic effects on responses to ionizing radiation, we not only propose this receptor as a therapeutic target but postulate that the biomarkers regulated in this study associated with radioprotection are potential mitigators of radiation-associated pathologies, including the onset of pulmonary disease.
Collapse
Affiliation(s)
- Elizabeth R Stirling
- Department of Cancer Biology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
| | - Katherine L Cook
- Department of Cancer Biology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Department of Surgery, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - David R Soto-Pantoja
- Department of Cancer Biology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Department of Surgery, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Department of Radiation Oncology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
| |
Collapse
|
32
|
Justice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA, Young K, Barata L, Deng X, Czajkowski J, Hadley D, Ngwa JS, Ahluwalia TS, Chu AY, Heard-Costa NL, Lim E, Perez J, Eicher JD, Kutalik Z, Xue L, Mahajan A, Renström F, Wu J, Qi Q, Ahmad S, Alfred T, Amin N, Bielak LF, Bonnefond A, Bragg J, Cadby G, Chittani M, Coggeshall S, Corre T, Direk N, Eriksson J, Fischer K, Gorski M, Neergaard Harder M, Horikoshi M, Huang T, Huffman JE, Jackson AU, Justesen JM, Kanoni S, Kinnunen L, Kleber ME, Komulainen P, Kumari M, Lim U, Luan J, Lyytikäinen LP, Mangino M, Manichaikul A, Marten J, Middelberg RPS, Müller-Nurasyid M, Navarro P, Pérusse L, Pervjakova N, Sarti C, Smith AV, Smith JA, Stančáková A, Strawbridge RJ, Stringham HM, Sung YJ, Tanaka T, Teumer A, Trompet S, van der Laan SW, van der Most PJ, Van Vliet-Ostaptchouk JV, Vedantam SL, Verweij N, Vink JM, Vitart V, Wu Y, Yengo L, Zhang W, Hua Zhao J, Zimmermann ME, Zubair N, Abecasis GR, Adair LS, Afaq S, Afzal U, Bakker SJL, Bartz TM, Beilby J, Bergman RN, Bergmann S, Biffar R, Blangero J, Boerwinkle E, Bonnycastle LL, Bottinger E, Braga D, Buckley BM, Buyske S, Campbell H, et alJustice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA, Young K, Barata L, Deng X, Czajkowski J, Hadley D, Ngwa JS, Ahluwalia TS, Chu AY, Heard-Costa NL, Lim E, Perez J, Eicher JD, Kutalik Z, Xue L, Mahajan A, Renström F, Wu J, Qi Q, Ahmad S, Alfred T, Amin N, Bielak LF, Bonnefond A, Bragg J, Cadby G, Chittani M, Coggeshall S, Corre T, Direk N, Eriksson J, Fischer K, Gorski M, Neergaard Harder M, Horikoshi M, Huang T, Huffman JE, Jackson AU, Justesen JM, Kanoni S, Kinnunen L, Kleber ME, Komulainen P, Kumari M, Lim U, Luan J, Lyytikäinen LP, Mangino M, Manichaikul A, Marten J, Middelberg RPS, Müller-Nurasyid M, Navarro P, Pérusse L, Pervjakova N, Sarti C, Smith AV, Smith JA, Stančáková A, Strawbridge RJ, Stringham HM, Sung YJ, Tanaka T, Teumer A, Trompet S, van der Laan SW, van der Most PJ, Van Vliet-Ostaptchouk JV, Vedantam SL, Verweij N, Vink JM, Vitart V, Wu Y, Yengo L, Zhang W, Hua Zhao J, Zimmermann ME, Zubair N, Abecasis GR, Adair LS, Afaq S, Afzal U, Bakker SJL, Bartz TM, Beilby J, Bergman RN, Bergmann S, Biffar R, Blangero J, Boerwinkle E, Bonnycastle LL, Bottinger E, Braga D, Buckley BM, Buyske S, Campbell H, Chambers JC, Collins FS, Curran JE, de Borst GJ, de Craen AJM, de Geus EJC, Dedoussis G, Delgado GE, den Ruijter HM, Eiriksdottir G, Eriksson AL, Esko T, Faul JD, Ford I, Forrester T, Gertow K, Gigante B, Glorioso N, Gong J, Grallert H, Grammer TB, Grarup N, Haitjema S, Hallmans G, Hamsten A, Hansen T, Harris TB, Hartman CA, Hassinen M, Hastie ND, Heath AC, Hernandez D, Hindorff L, Hocking LJ, Hollensted M, Holmen OL, Homuth G, Jan Hottenga J, Huang J, Hung J, Hutri-Kähönen N, Ingelsson E, James AL, Jansson JO, Jarvelin MR, Jhun MA, Jørgensen ME, Juonala M, Kähönen M, Karlsson M, Koistinen HA, Kolcic I, Kolovou G, Kooperberg C, Krämer BK, Kuusisto J, Kvaløy K, Lakka TA, Langenberg C, Launer LJ, Leander K, Lee NR, Lind L, Lindgren CM, Linneberg A, Lobbens S, Loh M, Lorentzon M, Luben R, Lubke G, Ludolph-Donislawski A, Lupoli S, Madden PAF, Männikkö R, Marques-Vidal P, Martin NG, McKenzie CA, McKnight B, Mellström D, Menni C, Montgomery GW, Musk AW(B, Narisu N, Nauck M, Nolte IM, Oldehinkel AJ, Olden M, Ong KK, Padmanabhan S, Peyser PA, Pisinger C, Porteous DJ, Raitakari OT, Rankinen T, Rao DC, Rasmussen-Torvik LJ, Rawal R, Rice T, Ridker PM, Rose LM, Bien SA, Rudan I, Sanna S, Sarzynski MA, Sattar N, Savonen K, Schlessinger D, Scholtens S, Schurmann C, Scott RA, Sennblad B, Siemelink MA, Silbernagel G, Slagboom PE, Snieder H, Staessen JA, Stott DJ, Swertz MA, Swift AJ, Taylor KD, Tayo BO, Thorand B, Thuillier D, Tuomilehto J, Uitterlinden AG, Vandenput L, Vohl MC, Völzke H, Vonk JM, Waeber G, Waldenberger M, Westendorp RGJ, Wild S, Willemsen G, Wolffenbuttel BHR, Wong A, Wright AF, Zhao W, Zillikens MC, Baldassarre D, Balkau B, Bandinelli S, Böger CA, Boomsma DI, Bouchard C, Bruinenberg M, Chasman DI, Chen YD, Chines PS, Cooper RS, Cucca F, Cusi D, Faire UD, Ferrucci L, Franks PW, Froguel P, Gordon-Larsen P, Grabe HJ, Gudnason V, Haiman CA, Hayward C, Hveem K, Johnson AD, Wouter Jukema J, Kardia SLR, Kivimaki M, Kooner JS, Kuh D, Laakso M, Lehtimäki T, Marchand LL, März W, McCarthy MI, Metspalu A, Morris AP, Ohlsson C, Palmer LJ, Pasterkamp G, Pedersen O, Peters A, Peters U, Polasek O, Psaty BM, Qi L, Rauramaa R, Smith BH, Sørensen TIA, Strauch K, Tiemeier H, Tremoli E, van der Harst P, Vestergaard H, Vollenweider P, Wareham NJ, Weir DR, Whitfield JB, Wilson JF, Tyrrell J, Frayling TM, Barroso I, Boehnke M, Deloukas P, Fox CS, Hirschhorn JN, Hunter DJ, Spector TD, Strachan DP, van Duijn CM, Heid IM, Mohlke KL, Marchini J, Loos RJF, Kilpeläinen TO, Liu CT, Borecki IB, North KE, Cupples LA. Genome-wide meta-analysis of 241,258 adults accounting for smoking behaviour identifies novel loci for obesity traits. Nat Commun 2017; 8:14977. [PMID: 28443625 PMCID: PMC5414044 DOI: 10.1038/ncomms14977] [Show More Authors] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 02/15/2017] [Indexed: 02/07/2023] Open
Abstract
Few genome-wide association studies (GWAS) account for environmental exposures, like smoking, potentially impacting the overall trait variance when investigating the genetic contribution to obesity-related traits. Here, we use GWAS data from 51,080 current smokers and 190,178 nonsmokers (87% European descent) to identify loci influencing BMI and central adiposity, measured as waist circumference and waist-to-hip ratio both adjusted for BMI. We identify 23 novel genetic loci, and 9 loci with convincing evidence of gene-smoking interaction (GxSMK) on obesity-related traits. We show consistent direction of effect for all identified loci and significance for 18 novel and for 5 interaction loci in an independent study sample. These loci highlight novel biological functions, including response to oxidative stress, addictive behaviour, and regulatory functions emphasizing the importance of accounting for environment in genetic analyses. Our results suggest that tobacco smoking may alter the genetic susceptibility to overall adiposity and body fat distribution.
Collapse
Affiliation(s)
- Anne E. Justice
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Misa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Virginia A. Fisher
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Kristin Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Llilda Barata
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Xuan Deng
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Jacek Czajkowski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - David Hadley
- Population Health Research Institute, St. George's, University of London, London, SW17 0RE, UK
- TransMed Systems, Inc., Cupertino, California 95014, USA
| | - Julius S. Ngwa
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore Maryland, USA
| | - Tarunveer S. Ahluwalia
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Audrey Y. Chu
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
| | - Nancy L. Heard-Costa
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Jeremiah Perez
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - John D. Eicher
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Luting Xue
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Frida Renström
- Department of Biobank Research, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
| | - Joseph Wu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Shafqat Ahmad
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Tamuno Alfred
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015GE, The Netherlands
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Amelie Bonnefond
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Jennifer Bragg
- Internal Medicine - Nephrology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Australia
| | - Martina Chittani
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Scott Coggeshall
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
| | - Tanguy Corre
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Nese Direk
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Dokuz Eylul University, Izmir, Turkey
| | - Joel Eriksson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Mathias Gorski
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Marie Neergaard Harder
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Momoko Horikoshi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Tao Huang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Epidemiology Domain, Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Jennifer E. Huffman
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Anne U. Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Johanne Marie Justesen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Leena Kinnunen
- Department of Health, National Institute for Health and Welfare, Helsinki, FI-00271 Finland
| | - Marcus E. Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Meena Kumari
- ISER, University of Essex, Colchester CO43SQ, UK
- Department of Epidemiology and Public Health, UCL, London, WC1E 6BT, UK
| | - Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St. Thomas' Foundation Trust, London, UK
| | - Ani Manichaikul
- Center for Public Health Genomics and Biostatistics Section, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Jonathan Marten
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Rita P. S. Middelberg
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Louis Pérusse
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Canada
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - Natalia Pervjakova
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Cinzia Sarti
- Department of Social and Health Care, City of Helsinki, Helsinki, Finland
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Alena Stančáková
- Department of Medicine, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Rona J. Strawbridge
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Heather M. Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore Maryland, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Germany
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, The Netherlands
| | - Sander W. van der Laan
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Peter J. van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | | | - Sailaja L. Vedantam
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Jacqueline M. Vink
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Loic Yengo
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Weihua Zhang
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Martina E. Zimmermann
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Niha Zubair
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Gonçalo R. Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Linda S. Adair
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Saima Afaq
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Uzma Afzal
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Stephan J. L. Bakker
- Department of Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Traci M. Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington 98101, USA
| | - John Beilby
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- PathWest Laboratory Medicine of WA, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboraty Medicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia 6009, Australia
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Reiner Biffar
- Clinic for Prosthetic Dentistry, Gerostomatology and Material Science, University Medicine Greifswald, Germany
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eric Boerwinkle
- Human Genetics Center, The University of Texas Health Science Center, PO Box 20186, Houston, Texas 77225, USA
| | - Lori L. Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Erwin Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Daniele Braga
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Ireland
| | - Steve Buyske
- Department of Genetics, Rutgers University, Piscataway, New Jersey 08854, USA
- Department of Statistics and Biostatistics, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Harry Campbell
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - John C. Chambers
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Francis S. Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Joanne E. Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Gert J. de Borst
- Department of Vascular Surgery, Division of Surgical Specialties, UMC Utrecht, The Netherlands
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, The Netherlands
| | - Eco J. C. de Geus
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- EMGO+ Institute Vrije Universiteit & Vrije Universiteit Medical Center
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Graciela E. Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | | | - Anna L. Eriksson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, UK
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15 Jamaica
| | - Karl Gertow
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Bruna Gigante
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU-University of Sassari
| | - Jian Gong
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Tanja B. Grammer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Saskia Haitjema
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Section for Nutritional Research, Umeå University, Umeå, Sweden
| | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Catharina A. Hartman
- Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Nicholas D. Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Andrew C. Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Lucia Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Lynne J. Hocking
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Mette Hollensted
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Germany
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jie Huang
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Joseph Hung
- School of Medicine and Pharmacology, The University of Western Australia, 25 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Cardiovascular Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, Tampere 33521, Finland
- Department of Pediatrics, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, 751 85, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
- Science for Life Laboratory, Uppsala University, Uppsala, 750 85, Sweden
| | - Alan L. James
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- School of Medicine and Pharmacology, The University of Western Australia, 25 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC–PHE Centre for Environment & Health, School of Public Health, Imperial College London, UK
- Center for Life Course Epidemiology, Faculty of Medicine, University of OuluP.O.Box 5000, FI-90014, Finland
- Biocenter Oulu, University of Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Kajaanintie 50, P.O.Box 20, FI-90220, 90029 Oulu, Finland
| | - Min A. Jhun
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Markus Juonala
- Department of Medicine, University of Turku, Turku 20520 Finland
- Division of Medicine, Turku University Hospital, Turku 20521, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33521, Finland
- Department of Clinical Physiology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Orthopedics and Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Heikki A. Koistinen
- Department of Health, National Institute for Health and Welfare, Helsinki, FI-00271 Finland
- Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, FI-00029 Finland
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, FI-00290 Finland
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, Croatia
| | - Genovefa Kolovou
- Department of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Bernhard K. Krämer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Kirsti Kvaløy
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio Campus, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Karin Leander
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nanette R. Lee
- USC-Office of Population Studies Foundation, Inc., University of San Carlos, Cebu City 6000, Philippines
- Department of Anthropology, Sociology and History, University of San Carlos, Cebu City 6000, Philippines
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala 751 85, Sweden
| | - Cecilia M. Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Li Ka Shing Centre for Health Information and Discovery, The Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, the Capital Region of Denmark, Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephane Lobbens
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Marie Loh
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Robert Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Gitta Lubke
- Department of Psychology, University of Notre Dame, Notre Dame, USA
| | - Anja Ludolph-Donislawski
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Sara Lupoli
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Pamela A. F. Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Reija Männikkö
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Nicholas G. Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Colin A. McKenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15 Jamaica
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Dan Mellström
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Grant W. Montgomery
- Molecular Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - AW (Bill) Musk
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- School of Population Health, The University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Narisu Narisu
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Germany
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Albertine J. Oldehinkel
- Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Matthias Olden
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Sandosh Padmanabhan
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Charlotta Pisinger
- Research Center for Prevention and Health, Glostrup Hospital, Glostrup Denmark
- Department of Public Health, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - David J. Porteous
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20521, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laura J. Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rajesh Rawal
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Treva Rice
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
| | - Stephanie A. Bien
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Igor Rudan
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, 09042, Monserrato, Italy
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, UK
| | - Kai Savonen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Salome Scholtens
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Bengt Sennblad
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Marten A. Siemelink
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Günther Silbernagel
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Austria
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Jan A. Staessen
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Science , University of Leuven, Campus Sint Rafael, Kapucijnenvoer 35, Leuven; Belgium
- R&D VitaK Group, Maastricht University, Brains Unlimited Building, Oxfordlaan 55, Maastricht, The Netherlands
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, UK
| | - Morris A. Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Amy J. Swift
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Kent D. Taylor
- Center for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor/UCLA Medical Center, Torrance, California, USA
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California, USA
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University of Chicago, Maywood, Illinois 61053, USA
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Dorothee Thuillier
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Jaakko Tuomilehto
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
- Department of Neurosciences and Preventive Medicine, Danube-University Krems, 3500 Krems, Austria
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Andre G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
- School of Nutrition, Université Laval, Québec, Canada
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Germany
| | - Judith M. Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Gérard Waeber
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - R. G. J. Westendorp
- Department of Public Health, and Center for Healthy Ageing, University of Copenhagen, Denmark
| | - Sarah Wild
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Bruce H. R. Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, 33 Bedford Place, London, WC1B 5JU, UK
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Damiano Baldassarre
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Carsten A. Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Dorret I. Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Marcel Bruinenberg
- Lifelines Cohort Study, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Yii-DerIda Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics, Harbor-UCLA, Torrance, California 90502, USA
| | - Peter S. Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University of Chicago, Maywood, Illinois 61053, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, 09042, Monserrato, Italy
- Dipartimento di Scienze Biomediche, Universita' degli Studi di Sassari, Sassari, Italy
| | - Daniele Cusi
- Sanipedia srl, Bresso (Milano), Italy and Institute of Biomedical Technologies National Centre of Research Segrate, Milano, Italy
| | - Ulf de Faire
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore Maryland, USA
| | - Paul W. Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
| | - Philippe Froguel
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill North Carolina, 27516, USA
| | - Hans- Jörgen Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Christopher A. Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, 90089, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, UCL, London, WC1E 6BT, UK
| | - Jaspal S. Kooner
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
- Faculty of Med, National Heart & Lung Institute, Cardiovascular Science, Hammersmith Campus, Hammersmith Hospital, Hammersmith Campus, Imperial College London, UK
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing at UCL, 33 Bedford Place, London, WC1B 5JU, UK
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
| | - Winfried März
- Synlab Academy, Synlab Services GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lyle J. Palmer
- School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Hematology, Division Laboratories & Pharmacy, UMC Utrecht, The Netherlands
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Ozren Polasek
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
- Department of Public Health, Faculty of Medicine, University of Split, Croatia
| | - Bruce M. Psaty
- Department of Medicine, University of Washington, Seattle, Washington 98195, USA
- Department of Epidemiology, University of Washington, Seattle, Washington 98101, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington 98101, USA
| | - Lu Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Blair H. Smith
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4RB, Scotland
| | - Thorkild I. A. Sørensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Epidemiology (formerly Institute of Preventive Medicine), Bispebjerg and Frederiksberg Hospital (2000 Frederiksberg), The Capital Region, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, Bristol University, Bristol, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Henning Tiemeier
- Department of Psychiatry Erasmus Medical Center, Rotterdam, The Netherlands
| | - Elena Tremoli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Henrik Vestergaard
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - John B. Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, RILD Building University of Exeter, Exeter, EX2 5DW, UK
- European Centre for Environment and Human Health, University of Exeter Medical School, The Knowledge Spa, Truro TR1 3HD, UK
| | - Timothy M. Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Level 4, Institute of Metabolic Science Box 289 Addenbrooke's Hospital Cambridge CB2 OQQ, UK
- University of Cambridge Metabolic Research Laboratories, Level 4, Institute of Metabolic Science Box 289 Addenbrooke's Hospital Cambridge CB2 OQQ, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Panagiotis Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Caroline S. Fox
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
| | - Joel N. Hirschhorn
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Department of Genetics, Harvard Medical School, Boston Massachusetts 02115, USA
| | - David J. Hunter
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - David P. Strachan
- Population Health Research Institute, St. George's, University of London, London, SW17 0RE, UK
- Division of Population Health Sciences and Education, St George's, University of London, London SW17 0RE, UK
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015GE, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA). Leiden, The Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
- Mount Sinai School of Medicine, New York 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Tuomas O. Kilpeläinen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
- Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
| |
Collapse
|
33
|
Ried JS, Jeff M. J, Chu AY, Bragg-Gresham JL, van Dongen J, Huffman JE, Ahluwalia TS, Cadby G, Eklund N, Eriksson J, Esko T, Feitosa MF, Goel A, Gorski M, Hayward C, Heard-Costa NL, Jackson AU, Jokinen E, Kanoni S, Kristiansson K, Kutalik Z, Lahti J, Luan J, Mägi R, Mahajan A, Mangino M, Medina-Gomez C, Monda KL, Nolte IM, Pérusse L, Prokopenko I, Qi L, Rose LM, Salvi E, Smith MT, Snieder H, Stančáková A, Ju Sung Y, Tachmazidou I, Teumer A, Thorleifsson G, van der Harst P, Walker RW, Wang SR, Wild SH, Willems SM, Wong A, Zhang W, Albrecht E, Couto Alves A, Bakker SJL, Barlassina C, Bartz TM, Beilby J, Bellis C, Bergman RN, Bergmann S, Blangero J, Blüher M, Boerwinkle E, Bonnycastle LL, Bornstein SR, Bruinenberg M, Campbell H, Chen YDI, Chiang CWK, Chines PS, Collins FS, Cucca F, Cupples LA, D'Avila F, de Geus EJ.C, Dedoussis G, Dimitriou M, Döring A, Eriksson JG, Farmaki AE, Farrall M, Ferreira T, Fischer K, Forouhi NG, Friedrich N, Gjesing AP, Glorioso N, Graff M, Grallert H, Grarup N, Gräßler J, Grewal J, Hamsten A, Harder MN, Hartman CA, Hassinen M, Hastie N, Hattersley AT, Havulinna AS, Heliövaara M, Hillege H, Hofman A, Holmen O, et alRied JS, Jeff M. J, Chu AY, Bragg-Gresham JL, van Dongen J, Huffman JE, Ahluwalia TS, Cadby G, Eklund N, Eriksson J, Esko T, Feitosa MF, Goel A, Gorski M, Hayward C, Heard-Costa NL, Jackson AU, Jokinen E, Kanoni S, Kristiansson K, Kutalik Z, Lahti J, Luan J, Mägi R, Mahajan A, Mangino M, Medina-Gomez C, Monda KL, Nolte IM, Pérusse L, Prokopenko I, Qi L, Rose LM, Salvi E, Smith MT, Snieder H, Stančáková A, Ju Sung Y, Tachmazidou I, Teumer A, Thorleifsson G, van der Harst P, Walker RW, Wang SR, Wild SH, Willems SM, Wong A, Zhang W, Albrecht E, Couto Alves A, Bakker SJL, Barlassina C, Bartz TM, Beilby J, Bellis C, Bergman RN, Bergmann S, Blangero J, Blüher M, Boerwinkle E, Bonnycastle LL, Bornstein SR, Bruinenberg M, Campbell H, Chen YDI, Chiang CWK, Chines PS, Collins FS, Cucca F, Cupples LA, D'Avila F, de Geus EJ.C, Dedoussis G, Dimitriou M, Döring A, Eriksson JG, Farmaki AE, Farrall M, Ferreira T, Fischer K, Forouhi NG, Friedrich N, Gjesing AP, Glorioso N, Graff M, Grallert H, Grarup N, Gräßler J, Grewal J, Hamsten A, Harder MN, Hartman CA, Hassinen M, Hastie N, Hattersley AT, Havulinna AS, Heliövaara M, Hillege H, Hofman A, Holmen O, Homuth G, Hottenga JJ, Hui J, Husemoen LL, Hysi PG, Isaacs A, Ittermann T, Jalilzadeh S, James AL, Jørgensen T, Jousilahti P, Jula A, Marie Justesen J, Justice AE, Kähönen M, Karaleftheri M, Tee Khaw K, Keinanen-Kiukaanniemi SM, Kinnunen L, Knekt PB, Koistinen HA, Kolcic I, Kooner IK, Koskinen S, Kovacs P, Kyriakou T, Laitinen T, Langenberg C, Lewin AM, Lichtner P, Lindgren CM, Lindström J, Linneberg A, Lorbeer R, Lorentzon M, Luben R, Lyssenko V, Männistö S, Manunta P, Leach IM, McArdle WL, Mcknight B, Mohlke KL, Mihailov E, Milani L, Mills R, Montasser ME, Morris AP, Müller G, Musk AW, Narisu N, Ong KK, Oostra BA, Osmond C, Palotie A, Pankow JS, Paternoster L, Penninx BW, Pichler I, Pilia MG, Polašek O, Pramstaller PP, Raitakari OT, Rankinen T, Rao DC, Rayner NW, Ribel-Madsen R, Rice TK, Richards M, Ridker PM, Rivadeneira F, Ryan KA, Sanna S, Sarzynski MA, Scholtens S, Scott RA, Sebert S, Southam L, Sparsø TH, Steinthorsdottir V, Stirrups K, Stolk RP, Strauch K, Stringham HM, Swertz MA, Swift AJ, Tönjes A, Tsafantakis E, van der Most PJ, Van Vliet-Ostaptchouk JV, Vandenput L, Vartiainen E, Venturini C, Verweij N, Viikari JS, Vitart V, Vohl MC, Vonk JM, Waeber G, Widén E, Willemsen G, Wilsgaard T, Winkler TW, Wright AF, Yerges-Armstrong LM, Hua Zhao J, Carola Zillikens M, Boomsma DI, Bouchard C, Chambers JC, Chasman DI, Cusi D, Gansevoort RT, Gieger C, Hansen T, Hicks AA, Hu F, Hveem K, Jarvelin MR, Kajantie E, Kooner JS, Kuh D, Kuusisto J, Laakso M, Lakka TA, Lehtimäki T, Metspalu A, Njølstad I, Ohlsson C, Oldehinkel AJ, Palmer LJ, Pedersen O, Perola M, Peters A, Psaty BM, Puolijoki H, Rauramaa R, Rudan I, Salomaa V, Schwarz PEH, Shudiner AR, Smit JH, Sørensen TIA, Spector TD, Stefansson K, Stumvoll M, Tremblay A, Tuomilehto J, Uitterlinden AG, Uusitupa M, Völker U, Vollenweider P, Wareham NJ, Watkins H, Wilson JF, Zeggini E, Abecasis GR, Boehnke M, Borecki IB, Deloukas P, van Duijn CM, Fox C, Groop LC, Heid IM, Hunter DJ, Kaplan RC, McCarthy MI, North KE, O'Connell JR, Schlessinger D, Thorsteinsdottir U, Strachan DP, Frayling T, Hirschhorn JN, Müller-Nurasyid M, Loos RJF. A principal component meta-analysis on multiple anthropometric traits identifies novel loci for body shape. Nat Commun 2016; 7:13357. [PMID: 27876822 PMCID: PMC5114527 DOI: 10.1038/ncomms13357] [Show More Authors] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 09/21/2016] [Indexed: 01/15/2023] Open
Abstract
Large consortia have revealed hundreds of genetic loci associated with anthropometric traits, one trait at a time. We examined whether genetic variants affect body shape as a composite phenotype that is represented by a combination of anthropometric traits. We developed an approach that calculates averaged PCs (AvPCs) representing body shape derived from six anthropometric traits (body mass index, height, weight, waist and hip circumference, waist-to-hip ratio). The first four AvPCs explain >99% of the variability, are heritable, and associate with cardiometabolic outcomes. We performed genome-wide association analyses for each body shape composite phenotype across 65 studies and meta-analysed summary statistics. We identify six novel loci: LEMD2 and CD47 for AvPC1, RPS6KA5/C14orf159 and GANAB for AvPC3, and ARL15 and ANP32 for AvPC4. Our findings highlight the value of using multiple traits to define complex phenotypes for discovery, which are not captured by single-trait analyses, and may shed light onto new pathways.
Collapse
Affiliation(s)
- Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Janina Jeff M.
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Audrey Y. Chu
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Jennifer L. Bragg-Gresham
- Kidney Epidemiology and Cost Center, Internal Medicine-Nephrology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jenny van Dongen
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Tarunveer S. Ahluwalia
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- Steno Diabetes Center A/S, DK-2820 Gentofte, Denmark
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Ledreborg Allé 34, DK-2820 Copenhagen, Denmark
| | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Niina Eklund
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Joel Eriksson
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Tõnu Esko
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Mathias Gorski
- Department of Nephrology, University Hospital Regensburg, 93042 Regensburg, Germany
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Nancy L. Heard-Costa
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Anne U. Jackson
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Eero Jokinen
- Hospital for Children and Adolescents, University of Helsinki, FI-00290 Helsinki, Finland
| | - Stavroula Kanoni
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Kati Kristiansson
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Zoltán Kutalik
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, 1005, Switzerland
- Institute of Social and Preventive Medicine, University Hospital Lausanne (CHUV), 1010 Lausanne, Switzerland
| | - Jari Lahti
- Folkhälsan Research Centre, FI-00290 Helsinki, Finland
- Institute of Behavioural Sciences, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Carolina Medina-Gomez
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Keri L. Monda
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- The Center for Observational Research, Amgen Inc., Thousand Oaks, California 91320-1799, USA
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Louis Pérusse
- Department of Kinesiology, Laval University, Québec, Québec, Canada G1V 0A6
- Institute of Nutrition and Functional Foods, Laval University, Québec, Québec, Canada G1V 0A6
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London W12 0NN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Lu Qi
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Erika Salvi
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Megan T. Smith
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Alena Stančáková
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Ioanna Tachmazidou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | | | - Pim van der Harst
- Durrer Center for Cardiogenetic Research, Interuniversity Cardiology Institute Netherlands-Netherlands Heart Institute, 3501 DG Utrecht, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Ryan W. Walker
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Sophie R. Wang
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Divisions of Genetics and Endocrinology and Program in Genomics, Boston's Children's Hospital, Boston, Massachusetts 02115, USA
| | - Sarah H. Wild
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Sara M. Willems
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London W12 0NN, UK
| | - Stephan J. L. Bakker
- Department of Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Cristina Barlassina
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Traci M. Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
| | - John Beilby
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Claire Bellis
- Genomics Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4001, Australia
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research of Singapore, Singapore 138672, Singapore
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Sven Bergmann
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Department of Medical Genetics, University of Lausanne, Lausanne, 1005, Switzerland
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas 78520, USA
| | - Matthias Blüher
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Lori L. Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Stefan R. Bornstein
- Medical Faculty Carl Gustav Carus, Department of Medicine III, University of Dresden, 01307 Dresden, Germany
| | - Marcel Bruinenberg
- University of Groningen, University Medical Center Groningen, The LifeLines Cohort Study, 9700 RB Groningen, The Netherlands
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Yii-Der Ida Chen
- Los Angeles BioMedical Resesarch Institute at Harbor-UCLA Medical Center, Torrance, California 90502, USA
| | | | - Peter S. Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Francis S Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | | | - L Adrienne Cupples
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
| | - Francesca D'Avila
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Eco J .C. de Geus
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
- EMGO Institute for Health and Care Research, VU University Medical Center, 1081 BT Amsterdam, The Netherlands
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Maria Dimitriou
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Angela Döring
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Johan G. Eriksson
- Folkhälsan Research Centre, FI-00290 Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, FI-00014 Helsinki, Finland
| | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Anette Prior Gjesing
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU-University of Sassari, 7100 Sassari, Italy
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Niels Grarup
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jürgen Gräßler
- Department of Medicine III, Pathobiochemistry, Technische Universitaet, 01307 Dresden, Germany
| | - Jagvir Grewal
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
| | - Anders Hamsten
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine Solna, Atherosclerosis Research Unit, Karolinska Institutet, 17176 Stockholm 17176, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Marie Neergaard Harder
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Catharina A. Hartman
- University of Groningen, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, 9700 RB Groningen, The Netherlands
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
| | - Nicholas Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Andrew Tym Hattersley
- Institue of Biomedical & Clinical Science, University of Exeter, Barrack Road, Exeter EX2 5DW, UK
| | - Aki S. Havulinna
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Markku Heliövaara
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Hans Hillege
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Oddgeir Holmen
- Department of Public Health and General Practice, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Jennie Hui
- Pathwest Laboratory Medicine of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia 6009, Australia
- School of Population Health, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Lise Lotte Husemoen
- Research Centre for Prevention and Health, Glostrup Hospital, 2600 Glostrup, Denmark
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Aaron Isaacs
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Till Ittermann
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Shapour Jalilzadeh
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Alan L. James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Torben Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Faculty of Medicine, University of Aalborg, 9220 Aalborg, Denmark
- Research Centre for Prevention and Health, Capital Region of Denmark, DK2600 Glostrup, Denmark
| | - Pekka Jousilahti
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Antti Jula
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Johanne Marie Justesen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, FI-33521 Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, FI-33014 Tampere, Finland
| | | | - Kay Tee Khaw
- Clinical Gerontology Unit, Box 251, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK
| | - Sirkka M. Keinanen-Kiukaanniemi
- Faculty of Medicine, Institute of Health Sciences, University of Oulu, Oulu F1-90014, Finland
- Unit of General Practice, Oulu University Hospital, Oulu FI-90029, Finland
| | - Leena Kinnunen
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Paul B. Knekt
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Heikki A. Koistinen
- National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital,, 00029 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, 21000 Split, Croatia
| | | | - Seppo Koskinen
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
| | - Theodosios Kyriakou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Tomi Laitinen
- Kuopio University Hospital, 70029 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK
| | - Alexandra M. Lewin
- Department of Epidemiology and Biostatistics, MRC Health Protection Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London W12 0NN, UK
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Cecilia M. Lindgren
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- The Big Data Institute, University of Oxford, Oxford OX3 7LJ, UK
| | - Jaana Lindström
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup Hospital, 2600 Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, 2600 Glostrup, Denmark
| | - Roberto Lorbeer
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Mattias Lorentzon
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Robert Luben
- Strangeways Research Laboratory Wort's Causeway, Cambridge CB1 8RN, UK
| | - Valeriya Lyssenko
- Steno Diabetes Center A/S, DK-2820 Gentofte, Denmark
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, 221 00 Malmö, Sweden
| | - Satu Männistö
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Paolo Manunta
- Chair of Nephrology, Università Vita Salute San Raffaele and Genomics of Renal Diseases and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan 20139, Italy
| | - Irene Mateo Leach
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol BS82BN, UK
| | - Barbara Mcknight
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
- Divison of Public Health Sciences, Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Lili Milani
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | | | - May E. Montasser
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GA, UK
| | - Gabriele Müller
- Center for Evidence Based Healthcare, University of Dresden, Medical Faculty Carl Gustav Carus, Dresden, 01307, Germany
| | - Arthur W. Musk
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, West Australia 6009, Australia
| | - Narisu Narisu
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ben A. Oostra
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
- Massachusetts General Hospital, Center for Human Genetic Research, Psychiatric and Neurodevelopmental Genetics Unit, Boston, Massachusetts 02114, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota 55455-0381, USA
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Brenda W. Penninx
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, AJ Ernstraat 1887, 1081 HL Amsterdam, The Netherlands
| | - Irene Pichler
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
| | - Maria G. Pilia
- Istituto di Ricerca Genetica e Biomedica, CNR, 9042 Monserrato, Italy
| | - Ozren Polašek
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
- Department of Public Health, Faculty of Medicine, University of Split, 21000 Split, Croatia
| | - Peter P. Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
- Department of Neurology, University of Lübeck, 23562 Lübeck, Germany
- Department of Neurology, General Central Hospital, 39100 Bolzano, Italy
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, FI-20521 Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, FI-20520 Turku, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Nigel W. Rayner
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1HH, UK
| | - Rasmus Ribel-Madsen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Marcus Richards
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Kathy A. Ryan
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica, CNR, 9042 Monserrato, Italy
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - Salome Scholtens
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Sylvain Sebert
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London W12 0NN, UK
- Biocenter Oulu, University of Oulu, Oulu FI-90014, Finland
- Center For Life-Course Health Research, University of Oulu, FI-90014 Oulu, Finland
| | - Lorraine Southam
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Thomas Hempel Sparsø
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | | | - Kathleen Stirrups
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ronald P. Stolk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Heather M. Stringham
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Morris A. Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Amy J. Swift
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Anke Tönjes
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | | | - Peter J. van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Jana V. Van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Liesbeth Vandenput
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Erkki Vartiainen
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Jorma S. Viikari
- Department of Medicine, University of Turku, FI-20521 Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Laval University, Québec, Québec, Canada G1V 0A6
- School of Nutrition, Laval University, Québec, Québec, Canada G1V 0A6
| | - Judith M. Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Gérard Waeber
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Tom Wilsgaard
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
| | - Laura M. Yerges-Armstrong
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - M. Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Dorret I. Boomsma
- Department of Biological Psychology, VU University, 1081BT Amsterdam, The Netherlands
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - John C. Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Daniele Cusi
- Department of Health Sciences, University of Milano at San Paolo Hospital, 20139 Milano, Italy
- Filarete Foundation, Genomic and Bioinformatics Unit, Milano 20139, Italy
| | - Ron T. Gansevoort
- Department of Medicine, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, Netherlands
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Torben Hansen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern Denmark, 5000 Odense, Denmark
| | - Andrew A. Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), 39100 Bolzano, Italy
- Affiliated Institute of the University of Lübeck, 23562 Lübeck, Germany
| | - Frank Hu
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Kristian Hveem
- Department of Public Health and General Practice, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Marjo-Riitta Jarvelin
- Biocenter Oulu, University of Oulu, Oulu FI-90014, Finland
- Unit of Primary Care, Oulu University Hospital, 90029 OYS Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC–PHE Centre for Environment & Health, School of Public Health, Imperial College London W12 0NN, UK
- Faculty of Medicine, Center for Life Course Epidemiology, University of Oulu, P.O.Box 5000, FI-90014 Oulu, Finland
| | - Eero Kajantie
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Children's Hospital, Helsinki University Hospital and University of Helsinki, FI-00029 Helsinki, Finland
- Department of Obstetrics and Gynecology, MRC Oulu, Oulu University Hospital and University of Oulu, FI-90029 Oulu, Finland
| | - Jaspal S. Kooner
- Ealing Hospital NHS Trust, Middlesex UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London W12 0HS, UK
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK
| | - Diana Kuh
- MRC Unit for Lifelong Health & Ageing at UCL, London WC1B 5JU, UK
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
- Department of Physiology, Institute of Biomedicine, University of Eastern Finland, Kuopio Campus, 70210 Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, University of Tampere School of Medicine, FI-33014 Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, FI-33520 Tampere, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Inger Njølstad
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
- Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, 9037 Tromsø, Norway
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Albertine J. Oldehinkel
- University of Groningen, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, 9700 RB Groningen, The Netherlands
| | - Lyle J. Palmer
- School of Public Health, University of Adelaide, Adelaide, South Australia 5005, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Oluf Pedersen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Markus Perola
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00290 Helsinki, Finland
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, 80802 Munich, Germany
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, Washington 98101, USA
- Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Departments of Epidemiology and Health Services, University of Washington, Seattle, Washington 98101, USA
- Group Health Research Institute, Group Health Cooperative, Seatte, Washington 98101, USA
| | - Hannu Puolijoki
- South Ostrobothnia Central Hospital, Seinäjoki Fi-60220, Finland
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, 70100 Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, 70211 Kuopio, Finland
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Veikko Salomaa
- Department of Health, National Institute for Health and Welfare (THL), FI-00271 Helsinki, Finland
| | - Peter E. H. Schwarz
- Medical Faculty Carl Gustav Carus, Department of Medicine III, University of Dresden, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD), Dresden 01307, Germany
| | - Alan R. Shudiner
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
- Geriatric Research and Education Clinical Center, Vetrans Administration Medical Center, Baltimore, Maryland 21042, USA
| | - Jan H. Smit
- Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Center, AJ Ernstraat 1887, 1081 HL Amsterdam, The Netherlands
| | - Thorkild I. A. Sørensen
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, 2100 Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol BS82BN, UK
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, 2000 Frederiksberg, Denmark
| | - Timothy D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - Kari Stefansson
- deCODE Genetics, Amgen inc., 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Michael Stumvoll
- University of Leipzig, IFB Adiposity Diseases, 04103 Leipzig, Germany
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Angelo Tremblay
- Department of Kinesiology, Laval University, Québec, Québec, Canada G1V 0A6
| | - Jaakko Tuomilehto
- Diabetes Prevention Unit, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
- Centre for Vascular Prevention, Danube-University Krems, 3500 Krems, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Matti Uusitupa
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, 70211 Kuopio, Finland
- Research Unit, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Peter Vollenweider
- Department of Internal Medicine, University Hospital Lausanne (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, Scotland
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, EH8 9AG Teviot Place, Edinburgh, Scotland
| | - Eleftheria Zeggini
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton, Cambridge CB10 1SA, UK
| | - Goncalo R. Abecasis
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Michael Boehnke
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Wellcome Trust Sanger Institute, Human Genetics, Hinxton CB10 1HH, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Cornelia M. van Duijn
- Department of Epidemiology, Genetic Epidemiology Unit, Erasmus University Medical Center, 3015GE Rotterdam, The Netherlands
- Center for Medical Systems Biology, 2300 Leiden, The Netherlands
| | - Caroline Fox
- National Heart, Lung, and Blood Institute, the Framingham Heart Study, Framingham, Massachusetts 01702, USA
- Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Leif C. Groop
- Lund University Diabetes Centre and Department of Clinical Science, Diabetes & Endocrinology Unit, Lund University, 221 00 Malmö, Sweden
- Finnish Institute for Molecular Medicine (FIMM), Helsinki University, 00014 Helsinki, Finland
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, 93053 Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - David J. Hunter
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts 02115, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts 02115, USA
| | - Robert C. Kaplan
- Department of Epidemiology and Popualtion Health, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre, Oxford OX3 7LJ, UK
| | - Kari E. North
- Department of Epidemiology, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7400, USA
| | - Jeffrey R. O'Connell
- Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - David Schlessinger
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen inc., 101 Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - David P. Strachan
- Population Health Research Institute, St George's, University of London, London SW17 0RE, UK
| | - Timothy Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Joel N. Hirschhorn
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Divisions of Genetics and Endocrinology and Program in Genomics, Boston's Children's Hospital, Boston, Massachusetts 02115, USA
- Metabolism Initiative, Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partnersite Munich Heart Alliance, 80802 Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- The Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Genetics of Obesity and Related Metabolic Traits Program, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
34
|
Dietary fat overcomes the protective activity of thrombospondin-1 signaling in the Apc(Min/+) model of colon cancer. Oncogenesis 2016; 5:e230. [PMID: 27239962 PMCID: PMC4945754 DOI: 10.1038/oncsis.2016.37] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/14/2016] [Accepted: 04/16/2016] [Indexed: 12/17/2022] Open
Abstract
Thrombospondin 1 is a glycoprotein that regulates cellular phenotype through interactions with its cellular receptors and extracellular matrix-binding partners. Thrombospondin 1 locally regulates angiogenesis and inflammatory responses that contribute to colorectal carcinogenesis in ApcMin/+ mice. The ability of thrombospondin 1 to regulate responses of cells and tissues to a variety of stresses suggested that loss of thrombospondin 1 may also have broader systemic effects on metabolism to modulate carcinogenesis. ApcMin/+:Thbs1−/− mice exhibited decreased survival and higher tumor multiplicities in the small and large intestine relative to ApcMin/+ mice when fed a low (5%) fat western diet. However, the protective effect of endogenous thrombospondin 1 was lost when the mice were fed a western diet containing 21% fat. Biochemical profiles of liver tissue identified systemic metabolic changes accompanying the effects of thrombospondin 1 and dietary lipid intake on tumorigenesis. A high-fat western diet differentially regulated elements of amino acid, energy and lipid metabolism in ApcMin/+:Thbs1−/− mice relative to ApcMin/+:Thbs1+/+mice. Metabolic changes in ketone body and tricarboxylic acid cycle intermediates indicate functional interactions between Apc and thrombospondin 1 signaling that control mitochondrial function. The cumulative diet-dependent differential changes observed in ApcMin/+:Thbs1−/− versus ApcMin/+ mice include altered amino acid and lipid metabolism, mitochondrial dysfunction, eicosanoids and ketone body formation. This metabolic profile suggests that the protective role of thrombospondin 1 to decrease adenoma formation in ApcMin/+ mice results in part from improved mitochondrial function.
Collapse
|
35
|
Si YC, Miao WN, He JY, Wang XF, Wang YL, Ding WJ. Intricate interactions of obesity, intestinal flora and Toll-like receptors. Shijie Huaren Xiaohua Zazhi 2016; 24:2361-2367. [DOI: 10.11569/wcjd.v24.i15.2361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The rapidly increasing incidence of obesity has resulted in a severe public problem globally. Obesity is associated with subclinical inflammation, causing elevated levels of inflammatory cytokines, as well as disorders of the immune function, which are involved in the dysfunction of intestinal flora. Intestinal flora maintains a dynamic equilibrium with intestinal mucosal immunity. Obesity-related inflammation is mainly trigged by endoplasmic reticulum stress, Toll-like receptor 4 (TLR4) activation and changes of gut flora. Among them, TLR4 plays a central role in sensing intestinal pathogens and inducing mucosal immunity. On the other hand, metabolism, genetics, gut flora and immune state are integrally regulating the TLR function. In the present paper we explore the intricate interactions of obesity, intestinal flora and TLRs, in order to find novel targets for the treatment of obesity.
Collapse
|
36
|
Helsley RN, Sui Y, Park SH, Liu Z, Lee RG, Zhu B, Kern PA, Zhou C. Targeting IκB kinase β in Adipocyte Lineage Cells for Treatment of Obesity and Metabolic Dysfunctions. Stem Cells 2016; 34:1883-95. [PMID: 26991836 DOI: 10.1002/stem.2358] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/15/2016] [Indexed: 02/06/2023]
Abstract
IκB kinase β (IKKβ), a central coordinator of inflammation through activation of nuclear factor-κB, has been identified as a potential therapeutic target for the treatment of obesity-associated metabolic dysfunctions. In this study, we evaluated an antisense oligonucleotide (ASO) inhibitor of IKKβ and found that IKKβ ASO ameliorated diet-induced metabolic dysfunctions in mice. Interestingly, IKKβ ASO also inhibited adipocyte differentiation and reduced adiposity in high-fat (HF)-fed mice, indicating an important role of IKKβ signaling in the regulation of adipocyte differentiation. Indeed, CRISPR/Cas9-mediated genomic deletion of IKKβ in 3T3-L1 preadipocytes blocked these cells differentiating into adipocytes. To further elucidate the role of adipose progenitor IKKβ signaling in diet-induced obesity, we generated mice that selectively lack IKKβ in the white adipose lineage and confirmed the essential role of IKKβ in mediating adipocyte differentiation in vivo. Deficiency of IKKβ decreased HF-elicited adipogenesis in addition to reducing inflammation and protected mice from diet-induced obesity and insulin resistance. Further, pharmacological inhibition of IKKβ also blocked human adipose stem cell differentiation. Our findings establish IKKβ as a pivotal regulator of adipogenesis and suggest that overnutrition-mediated IKKβ activation serves as an initial signal that triggers adipose progenitor cell differentiation in response to HF feeding. Inhibition of IKKβ with antisense therapy may represent as a novel therapeutic approach to combat obesity and metabolic dysfunctions. Stem Cells 2016;34:1883-1895.
Collapse
Affiliation(s)
- Robert N Helsley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Yipeng Sui
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Zun Liu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Richard G Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Beibei Zhu
- Department of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Philip A Kern
- Department of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
37
|
Shi CX, Zhao MX, Shu XD, Xiong XQ, Wang JJ, Gao XY, Chen Q, Li YH, Kang YM, Zhu GQ. β-aminoisobutyric acid attenuates hepatic endoplasmic reticulum stress and glucose/lipid metabolic disturbance in mice with type 2 diabetes. Sci Rep 2016; 6:21924. [PMID: 26907958 PMCID: PMC4764829 DOI: 10.1038/srep21924] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/02/2016] [Indexed: 02/06/2023] Open
Abstract
β-aminoisobutyric acid (BAIBA) is a nature thymine catabolite, and contributes to exercise-induced protection from metabolic diseases. Here we show the therapeutical effects of BAIBA on hepatic endoplasmic reticulum (ER) stress and glucose/lipid metabolic disturbance in diabetes. Type 2 diabetes was induced by combined streptozotocin (STZ) and high-fat diet (HFD) in mice. Oral administration of BAIBA for 4 weeks reduced blood glucose and lipids levels, hepatic key enzymes of gluconeogenesis and lipogenesis expressions, attenuated hepatic insulin resistance and lipid accumulation, and improved insulin signaling in type 2 diabetic mice. BAIBA reduced hepatic ER stress and apoptosis in type 2 diabetic mice. Furthermore, BAIBA alleviated ER stress in human hepatocellular carcinoma (HepG2) cells with glucosamine-induced insulin resistance. Hepatic AMPK phosphorylation was reduced in STZ/HFD mice and glucosamine-treated HepG2 cells, which were restored by BAIBA treatment. The suppressive effects of BAIBA on glucosamine-induced ER stress were reversed by knockdown of AMPK with siRNA. In addition, BAIBA prevented thapsigargin- or tunicamycin-induced ER stress, and tunicamycin–induced apoptosis in HepG2 cells. These results indicate that BAIBA attenuates hepatic ER stress, apoptosis and glucose/lipid metabolic disturbance in mice with type 2 diabetes. AMPK signaling is involved to the role of BAIBA in attenuating ER stress.
Collapse
Affiliation(s)
- Chang-Xiang Shi
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming-Xia Zhao
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiao-Dong Shu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiao-Qing Xiong
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jue-Jin Wang
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xing-Ya Gao
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an 710061, China
| | - Guo-Qing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|