501
|
Sjögren syndrome/scleroderma autoantigen 1 is a direct Tankyrase binding partner in cancer cells. Commun Biol 2020; 3:123. [PMID: 32170109 PMCID: PMC7070046 DOI: 10.1038/s42003-020-0851-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/21/2020] [Indexed: 12/30/2022] Open
Abstract
Sjögren syndrome/scleroderma autoantigen 1 (SSSCA1) was first described as an auto-antigen over-expressed in Sjögren’s syndrome and in scleroderma patients. SSSCA1 has been linked to mitosis and centromere association and as a potential marker candidate in diverse solid cancers. Here we characterize SSSCA1 for the first time, to our knowledge, at the molecular, structural and subcellular level. We have determined the crystal structure of a zinc finger fold, a zinc ribbon domain type 2 (ZNRD2), at 2.3 Å resolution. We show that the C-terminal domain serves a dual function as it both behaves as the interaction site to Tankyrase 1 (TNKS1) and as a nuclear export signal. We identify TNKS1 as a direct binding partner of SSSCA1, map the binding site to TNKS1 ankyrin repeat cluster 2 (ARC2) and thus define a new binding sequence. We experimentally verify and map a new nuclear export signal sequence in SSSCA1. Perdreau-Dahl et al. systematically characterise Sjögren syndrome/scleroderma autoantigen 1 at the molecular, structural and subcellular level. They show that the C-terminal domain serves a dual function as it both behaves as the interaction site to Tankyrase 1 and as a nuclear export signal.
Collapse
|
502
|
van der Wijst MGP, de Vries DH, Groot HE, Trynka G, Hon CC, Bonder MJ, Stegle O, Nawijn MC, Idaghdour Y, van der Harst P, Ye CJ, Powell J, Theis FJ, Mahfouz A, Heinig M, Franke L. The single-cell eQTLGen consortium. eLife 2020; 9:e52155. [PMID: 32149610 PMCID: PMC7077978 DOI: 10.7554/elife.52155] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/03/2020] [Indexed: 12/17/2022] Open
Abstract
In recent years, functional genomics approaches combining genetic information with bulk RNA-sequencing data have identified the downstream expression effects of disease-associated genetic risk factors through so-called expression quantitative trait locus (eQTL) analysis. Single-cell RNA-sequencing creates enormous opportunities for mapping eQTLs across different cell types and in dynamic processes, many of which are obscured when using bulk methods. Rapid increase in throughput and reduction in cost per cell now allow this technology to be applied to large-scale population genetics studies. To fully leverage these emerging data resources, we have founded the single-cell eQTLGen consortium (sc-eQTLGen), aimed at pinpointing the cellular contexts in which disease-causing genetic variants affect gene expression. Here, we outline the goals, approach and potential utility of the sc-eQTLGen consortium. We also provide a set of study design considerations for future single-cell eQTL studies.
Collapse
Affiliation(s)
- MGP van der Wijst
- Department of Genetics, Oncode Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - DH de Vries
- Department of Genetics, Oncode Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - HE Groot
- Department of Cardiology, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - G Trynka
- Wellcome Sanger InstituteHinxtonUnited Kingdom
- Open TargetsHinxtonUnited Kingdom
| | - CC Hon
- RIKEN Center for Integrative Medical SciencesYokahamaJapan
| | - MJ Bonder
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ)HeidelbergGermany
- Genome Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
| | - O Stegle
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ)HeidelbergGermany
- Genome Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
| | - MC Nawijn
- Department of Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - Y Idaghdour
- Program in Biology, Public Health Research Center, New York University Abu DhabiAbu DhabiUnited Arab Emirates
| | - P van der Harst
- Department of Cardiology, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - CJ Ye
- Institute for Human Genetics, Bakar Computational Health Sciences Institute, Bakar ImmunoX Initiative, Department of Medicine, Department of Bioengineering and Therapeutic Sciences, Department of Epidemiology and Biostatistics, Chan Zuckerberg Biohub, University of California San FranciscoSan FranciscoUnited States
| | - J Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute, UNSW Cellular Genomics Futures Institute, University of New South WalesSydneyAustralia
| | - FJ Theis
- Institute of Computational Biology, Helmholtz Zentrum MünchenNeuherbergGermany
- Department of Mathematics, Technical University of MunichGarching bei MünchenGermany
| | - A Mahfouz
- Leiden Computational Biology Center, Leiden University Medical CenterLeidenNetherlands
- Delft Bioinformatics Lab, Delft University of TechnologyDelftNetherlands
| | - M Heinig
- Institute of Computational Biology, Helmholtz Zentrum MünchenNeuherbergGermany
- Department of Informatics, Technical University of MunichGarching bei MünchenGermany
| | - L Franke
- Department of Genetics, Oncode Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| |
Collapse
|
503
|
Li H, Khor CC, Fan J, Lv J, Yu C, Guo Y, Bian Z, Yang L, Millwood IY, Walters RG, Chen Y, Yuan JM, Yang Y, Hu C, Chen J, Chen Z, Koh WP, Huang T, Li L. Genetic risk, adherence to a healthy lifestyle, and type 2 diabetes risk among 550,000 Chinese adults: results from 2 independent Asian cohorts. Am J Clin Nutr 2020; 111:698-707. [PMID: 31974579 PMCID: PMC7049535 DOI: 10.1093/ajcn/nqz310] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Whether genetic susceptibility to type 2 diabetes is modified by a healthy lifestyle among Chinese remains unknown. OBJECTIVES The aim of the study was to determine whether genetic risk and adherence to a healthy lifestyle contribute independently to the risk of developing type 2 diabetes. METHODS We defined a lifestyle score using BMI, alcohol intake, smoking, physical activities, and diets in 461,030 participants from the China Kadoorie Biobank and 38,434 participants from the Singapore Chinese Health Study. A genetic risk score was constructed based on type 2 diabetes loci among 100,175 and 16,172 participants in each cohort, respectively. A Cox proportional-hazards model was used to estimate the interaction between genetic and lifestyle factors on the risk of type 2 diabetes. RESULTS In 2 independent Asian cohorts, we consistently found a healthy lifestyle (the bottom quintile of lifestyle score) was associated with a substantially lower risk of type 2 diabetes than an unhealthy lifestyle (the top quintile of lifestyle score) regardless of genetic risk. In those at a high genetic risk, the risk of type 2 diabetes was 57% lower among participants with a healthy lifestyle than among those with an unhealthy lifestyle in the pooled cohorts. Among participants at high genetic risk, the standardized 10-y incidence of type 2 diabetes was 7.11% in those with an unhealthy lifestyle vs. 2.45% in those with a healthy lifestyle. CONCLUSIONS In 2 independent cohorts involving 558,302 Chinese participants, we did not observe an interaction between genetics and lifestyle with type 2 diabetes risk, but our findings provide replicable evidence to show lifestyle factors and genetic factors were independently associated with the risk of type 2 diabetes. Within any genetic risk category, a healthy lifestyle was associated with a significantly lower risk of type 2 diabetes among the Chinese population.
Collapse
Affiliation(s)
- Haoxin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Chiea-Chuen Khor
- Genome Institute of Singapore, Singapore
- Singapore Eye Research Institute, Singapore
| | - Junning Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
- Peking University Institute of Environmental Medicine, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, China
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Beijing, China
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Robin G Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Yan Yang
- Huixian People's Hospital, Huixian, Henan, China
| | - Chen Hu
- NCDs Prevention and Control Department, Huixian CDC, Huixian, Henan, China
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Woon-Puay Koh
- Health Services and Systems Research, Duke-NUS Medical School, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
504
|
Weir GC, Gaglia J, Bonner-Weir S. Inadequate β-cell mass is essential for the pathogenesis of type 2 diabetes. Lancet Diabetes Endocrinol 2020; 8:249-256. [PMID: 32006519 PMCID: PMC7098467 DOI: 10.1016/s2213-8587(20)30022-x] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/19/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022]
Abstract
For patients with type 1 diabetes, it is accepted among the scientific community that there is a marked reduction in β-cell mass; however, with type 2 diabetes, there is disagreement as to whether this reduction in mass occurs in every case. Some have argued that β-cell mass in some patients with type 2 diabetes is normal and that the cause of the hyperglycaemia in these patients is a functional abnormality of insulin secretion. In this Personal View, we argue that a deficient β-cell mass is essential for the development of type 2 diabetes. The main point is that there are enormous (≥10 fold) variations in insulin sensitivity and insulin secretion in the general population, with a very close correlation between these two factors for any individual. Although β-cell mass cannot be accurately measured in living patients, it is highly likely that it too is highly correlated with insulin sensitivity and secretion. Thus, our argument is that a person with type 2 diabetes can have a β-cell mass that is the same as a person without type 2 diabetes, but because they are insulin resistant, the mass is inadequate and responsible for their diabetes. Because the abnormal insulin secretion of diabetes is caused by dysglycaemia and can be largely reversed with glycaemic control, it is a less serious problem than the reduction in β-cell mass, which is far more difficult to restore.
Collapse
Affiliation(s)
- Gordon C Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Jason Gaglia
- Section on Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Susan Bonner-Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
505
|
Eaaswarkhanth M, dos Santos ALC, Gokcumen O, Al-Mulla F, Thanaraj TA. Genome-Wide Selection Scan in an Arabian Peninsula Population Identifies a TNKS Haplotype Linked to Metabolic Traits and Hypertension. Genome Biol Evol 2020; 12:77-87. [PMID: 32068798 PMCID: PMC7093833 DOI: 10.1093/gbe/evaa033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the extreme and varying environmental conditions prevalent in the Arabian Peninsula, it has experienced several waves of human migrations following the out-of-Africa diaspora. Eventually, the inhabitants of the peninsula region adapted to the hot and dry environment. The adaptation and natural selection that shaped the extant human populations of the Arabian Peninsula region have been scarcely studied. In an attempt to explore natural selection in the region, we analyzed 662,750 variants in 583 Kuwaiti individuals. We searched for regions in the genome that display signatures of positive selection in the Kuwaiti population using an integrative approach in a conservative manner. We highlight a haplotype overlapping TNKS that showed strong signals of positive selection based on the results of the multiple selection tests conducted (integrated Haplotype Score, Cross Population Extended Haplotype Homozygosity, Population Branch Statistics, and log-likelihood ratio scores). Notably, the TNKS haplotype under selection potentially conferred a fitness advantage to the Kuwaiti ancestors for surviving in the harsh environment while posing a major health risk to present-day Kuwaitis.
Collapse
Affiliation(s)
| | - Andre Luiz Campelo dos Santos
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo
- Department of Archeology, Federal University of Pernambuco, Recife, Brazil
| | - Omer Gokcumen
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | |
Collapse
|
506
|
Jiang N, Lee S, Park T. Hierarchical structural component model for pathway analysis of common variants. BMC Med Genomics 2020; 13:26. [PMID: 32093692 PMCID: PMC7038534 DOI: 10.1186/s12920-019-0650-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have been widely used to identify phenotype-related genetic variants using many statistical methods, such as logistic and linear regression. However, GWAS-identified SNPs, as identified with stringent statistical significance, explain just a small portion of the overall estimated genetic heritability. To address this 'missing heritability' issue, gene- and pathway-based analysis, and biological mechanisms, have been used for many GWAS studies. However, many of these methods often neglect the correlation between genes and between pathways. METHODS We constructed a hierarchical component model that considers correlations both between genes and between pathways. Based on this model, we propose a novel pathway analysis method for GWAS datasets, Hierarchical structural Component Model for Pathway analysis of Common vAriants (HisCoM-PCA). HisCoM-PCA first summarizes the common variants of each gene, first at the gene-level, and then analyzes all pathways simultaneously by ridge-type penalization of both the gene and pathway effects on the phenotype. Statistical significance of the gene and pathway coefficients can be examined by permutation tests. RESULTS Using the simulation data set of Genetic Analysis Workshop 17 (GAW17), for both binary and continuous phenotypes, we showed that HisCoM-PCA well-controlled type I error, and had a higher empirical power compared to several other methods. In addition, we applied our method to a SNP chip dataset of KARE for four human physiologic traits: (1) type 2 diabetes; (2) hypertension; (3) systolic blood pressure; and (4) diastolic blood pressure. Those results showed that HisCoM-PCA could successfully identify signal pathways with superior statistical and biological significance. CONCLUSIONS Our approach has the advantage of providing an intuitive biological interpretation for associations between common variants and phenotypes, via pathway information, potentially addressing the missing heritability conundrum.
Collapse
Affiliation(s)
- Nan Jiang
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea
| | - Sungyoung Lee
- Center for Precision Medicine, Seoul National University Hospital, Seoul, 03080, Korea
| | - Taesung Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Korea.
- Department of Statistics, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
507
|
Guo X, Dai X, Zhou T, Wang H, Ni J, Xue J, Wang X. Mosaic loss of human Y chromosome: what, how and why. Hum Genet 2020; 139:421-446. [DOI: 10.1007/s00439-020-02114-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023]
|
508
|
Özdemir V, Arga KY, Aziz RK, Bayram M, Conley SN, Dandara C, Endrenyi L, Fisher E, Garvey CK, Hekim N, Kunej T, Şardaş S, Von Schomberg R, Yassin AS, Yılmaz G, Wang W. Digging Deeper into Precision/Personalized Medicine: Cracking the Sugar Code, the Third Alphabet of Life, and Sociomateriality of the Cell. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:62-80. [PMID: 32027574 DOI: 10.1089/omi.2019.0220] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Precision/personalized medicine is a hot topic in health care. Often presented with the motto "the right drug, for the right patient, at the right dose, and the right time," precision medicine is a theory for rational therapeutics as well as practice to individualize health interventions (e.g., drugs, food, vaccines, medical devices, and exercise programs) using biomarkers. Yet, an alien visitor to planet Earth reading the contemporary textbooks on diagnostics might think precision medicine requires only two biomolecules omnipresent in the literature: nucleic acids (e.g., DNA) and proteins, known as the first and second alphabet of biology, respectively. However, the precision/personalized medicine community has tended to underappreciate the third alphabet of life, the "sugar code" (i.e., the information stored in glycans, glycoproteins, and glycolipids). This article brings together experts in precision/personalized medicine science, pharmacoglycomics, emerging technology governance, cultural studies, contemporary art, and responsible innovation to critically comment on the sociomateriality of the three alphabets of life together. First, the current transformation of targeted therapies with personalized glycomedicine and glycan biomarkers is examined. Next, we discuss the reasons as to why unraveling of the sugar code might have lagged behind the DNA and protein codes. While social scientists have historically noted the importance of constructivism (e.g., how people interpret technology and build their values, hopes, and expectations into emerging technologies), life scientists relied on the material properties of technologies in explaining why some innovations emerge rapidly and are more popular than others. The concept of sociomateriality integrates these two explanations by highlighting the inherent entanglement of the social and the material contributions to knowledge and what is presented to us as reality from everyday laboratory life. Hence, we present a hypothesis based on a sociomaterial conceptual lens: because materiality and synthesis of glycans are not directly driven by a template, and thus more complex and open ended than sequencing of a finite length genome, social construction of expectations from unraveling of the sugar code versus the DNA code might have evolved differently, as being future-uncertain versus future-proof, respectively, thus potentially explaining the "sugar lag" in precision/personalized medicine diagnostics over the past decades. We conclude by introducing systems scientists, physicians, and biotechnology industry to the concept, practice, and value of responsible innovation, while glycomedicine and other emerging biomarker technologies (e.g., metagenomics and pharmacomicrobiomics) transition to applications in health care, ecology, pharmaceutical/diagnostic industries, agriculture, food, and bioengineering, among others.
Collapse
Affiliation(s)
- Vural Özdemir
- OMICS: A Journal of Integrative Biology, New Rochelle, New York.,Senior Advisor and Writer, Emerging Technology Governance and Responsible Innovation, Toronto, Ontario, Canada
| | - K Yalçın Arga
- Health Institutes of Turkey, Istanbul, Turkey.,Department of Bioengineering, Faculty of Engineering, Marmara University, İstanbul, Turkey
| | - Ramy K Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.,The Center for Genome and Microbiome Research, Cairo University, Cairo, Egypt
| | - Mustafa Bayram
- Department of Food Engineering, Faculty of Engineering, Gaziantep University, Gaziantep, Turkey
| | - Shannon N Conley
- STS Futures Lab, School of Integrated Sciences, James Madison University, Harrisonburg, Virginia
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology and Institute for Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Laszlo Endrenyi
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Erik Fisher
- School for the Future of Innovation in Society and the Consortium for Science, Policy and Outcomes, Arizona State University, Tempe, Arizona
| | - Colin K Garvey
- Stanford Institute for Human-Centered Artificial Intelligence, Stanford University, Palo Alto, California
| | - Nezih Hekim
- Department of Biochemistry, Faculty of Medicine, İstanbul Medipol University, İstanbul, Turkey
| | - Tanja Kunej
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Domzale, Slovenia
| | - Semra Şardaş
- Faculty of Pharmacy, İstinye University, İstanbul, Turkey
| | - Rene Von Schomberg
- Directorate General for Research and Innovation, European Commission, Brussel, Belgium.,Technical University Darmstadt, Darmstadt, Germany
| | - Aymen S Yassin
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.,The Center for Genome and Microbiome Research, Cairo University, Cairo, Egypt
| | - Gürçim Yılmaz
- Writer and Editor, Cultural Studies, and Curator of Contemporary Arts, İstanbul, Turkey
| | - Wei Wang
- Key Municipal Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| |
Collapse
|
509
|
Pharmacological enrichment of polygenic risk for precision medicine in complex disorders. Sci Rep 2020; 10:879. [PMID: 31964963 PMCID: PMC6972917 DOI: 10.1038/s41598-020-57795-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 01/03/2020] [Indexed: 12/29/2022] Open
Abstract
Individuals with complex disorders typically have a heritable burden of common variation that can be expressed as a polygenic risk score (PRS). While PRS has some predictive utility, it lacks the molecular specificity to be directly informative for clinical interventions. We therefore sought to develop a framework to quantify an individual’s common variant enrichment in clinically actionable systems responsive to existing drugs. This was achieved with a metric designated the pharmagenic enrichment score (PES), which we demonstrate for individual SNP profiles in a cohort of cases with schizophrenia. A large proportion of these had elevated PES in one or more of eight clinically actionable gene-sets enriched with schizophrenia associated common variation. Notable candidates targeting these pathways included vitamins, antioxidants, insulin modulating agents, and cholinergic drugs. Interestingly, elevated PES was also observed in individuals with otherwise low common variant burden. The biological saliency of PES profiles were observed directly through their impact on gene expression in a subset of the cohort with matched transcriptomic data, supporting our assertion that this gene-set orientated approach could integrate an individual’s common variant risk to inform personalised interventions, including drug repositioning, for complex disorders such as schizophrenia.
Collapse
|
510
|
Chen Z, Bai Y, Long X, Luo Q, Wen Z, Li Y, Huang S, Yan Y, Mo Z. Effects of Adiponectin on T2DM and Glucose Homeostasis: A Mendelian Randomization Study. Diabetes Metab Syndr Obes 2020; 13:1771-1784. [PMID: 32547139 PMCID: PMC7250315 DOI: 10.2147/dmso.s248352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/19/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE The associations of adiponectin with type 2 diabetes mellitus (T2DM), glucose homeostasis (including β-cell function index (HOMA-β), insulin resistance (HOMA-IR), fasting insulin (FI) and fasting glucose (FG)) have reported in epidemiological studies. However, the previous observational studies are prone to biases, such as reverse causation and residual confounding factors. Herein, a Mendelian Randomization (MR) study was conducted to determine whether causal effects exist among them. MATERIALS AND AND METHODS Two-sample MR analyses and multiple sensitivity analyses were performed using the summary data from the ADIPOGen consortium, MAGIC Consortium, and a meta-analysis of GWAS with a considerable sample of T2DM (62,892 cases and 596,424 controls of European ancestry). We got eight valid genetic variants to predict the causal effect among adiponectin and T2DM and glucose homeostasis after excluding the probable invalid or pleiotropic variants. RESULTS Adiponectin was not associated with T2DM (odds ratio (OR) = 1.004; 95% confidence interval (CI): 0.740, 1.363) when using MR Egger after removing the invalid SNPs, and the results were consistent when using the other four methods. Similar results existed among adiponectin and HOMA-β, HOMA-IR, FI, FG. CONCLUSION Our MR study revealed that adiponectin had no causal effect on T2DM and glucose homeostasis and that the associations among them in observational studies may be due to confounding factors.
Collapse
Affiliation(s)
- Zefeng Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Yulan Bai
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Xinyang Long
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Qianqian Luo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Zheng Wen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Yuanfan Li
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Yunkun Yan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- School of Public Health, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning530021, Guangxi, People’s Republic of China
- Guangxi Key Laboratory of Colleges and Universities, Nanning530021, Guangxi, People’s Republic of China
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning530021, Guangxi, People’s Republic of China
- Correspondence: Zengnan Mo Center for Genomic and Personalized Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning530021, Guangxi, People’s Republic of ChinaTel +86771-5353342 Email
| |
Collapse
|
511
|
Killion EA, Lu SC, Fort M, Yamada Y, Véniant MM, Lloyd DJ. Glucose-Dependent Insulinotropic Polypeptide Receptor Therapies for the Treatment of Obesity, Do Agonists = Antagonists? Endocr Rev 2020; 41:5568102. [PMID: 31511854 DOI: 10.1210/endrev/bnz002] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/03/2019] [Indexed: 12/19/2022]
Abstract
Glucose-dependent insulinotropic polypeptide receptor (GIPR) is associated with obesity in human genome-wide association studies. Similarly, mouse genetic studies indicate that loss of function alleles and glucose-dependent insulinotropic polypeptide overexpression both protect from high-fat diet-induced weight gain. Together, these data provide compelling evidence to develop therapies targeting GIPR for the treatment of obesity. Further, both antagonists and agonists alone prevent weight gain, but result in remarkable weight loss when codosed or molecularly combined with glucagon-like peptide-1 analogs preclinically. Here, we review the current literature on GIPR, including biology, human and mouse genetics, and pharmacology of both agonists and antagonists, discussing the similarities and differences between the 2 approaches. Despite opposite approaches being investigated preclinically and clinically, there may be viability of both agonists and antagonists for the treatment of obesity, and we expect this area to continue to evolve with new clinical data and molecular and pharmacological analyses of GIPR function.
Collapse
Affiliation(s)
- Elizabeth A Killion
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, California
| | - Shu-Chen Lu
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, California
| | - Madeline Fort
- Department of Comparative Biology and Safety Sciences, Amgen Research, Thousand Oaks, California
| | - Yuichiro Yamada
- Department of Endocrinology, Diabetes and Geriatric Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Murielle M Véniant
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, California
| | - David J Lloyd
- Department of Cardiometabolic Disorders, Amgen Research, Thousand Oaks, California
| |
Collapse
|
512
|
Park C, Jiang N, Park T. Pure additive contribution of genetic variants to a risk prediction model using propensity score matching: application to type 2 diabetes. Genomics Inform 2019; 17:e47. [PMID: 31896247 PMCID: PMC6944048 DOI: 10.5808/gi.2019.17.4.e47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/09/2019] [Indexed: 12/29/2022] Open
Abstract
The achievements of genome-wide association studies have suggested ways to predict diseases, such as type 2 diabetes (T2D), using single-nucleotide polymorphisms (SNPs). Most T2D risk prediction models have used SNPs in combination with demographic variables. However, it is difficult to evaluate the pure additive contribution of genetic variants to classically used demographic models. Since prediction models include some heritable traits, such as body mass index, the contribution of SNPs using unmatched case-control samples may be underestimated. In this article, we propose a method that uses propensity score matching to avoid underestimation by matching case and control samples, thereby determining the pure additive contribution of SNPs. To illustrate the proposed propensity score matching method, we used SNP data from the Korea Association Resources project and reported SNPs from the genome-wide association study catalog. We selected various SNP sets via stepwise logistic regression (SLR), least absolute shrinkage and selection operator (LASSO), and the elastic-net (EN) algorithm. Using these SNP sets, we made predictions using SLR, LASSO, and EN as logistic regression modeling techniques. The accuracy of the predictions was compared in terms of area under the receiver operating characteristic curve (AUC). The contribution of SNPs to T2D was evaluated by the difference in the AUC between models using only demographic variables and models that included the SNPs. The largest difference among our models showed that the AUC of the model using genetic variants with demographic variables could be 0.107 higher than that of the corresponding model using only demographic variables.
Collapse
Affiliation(s)
- Chanwoo Park
- Department of Statistics, Seoul National University, Seoul 08826, Korea
| | - Nan Jiang
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Korea
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul 08826, Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08826, Korea
- Corresponding author: E-mail:
| |
Collapse
|
513
|
Zhang H, Mo X, Qian Q, Zhou Z, Zhu Z, HuangFu X, Xu T, Wang A, Guo Z, Lei S, Zhang Y. Associations between potentially functional CORIN SNPs and serum corin levels in the Chinese Han population. BMC Genet 2019; 20:99. [PMID: 31856714 PMCID: PMC6923953 DOI: 10.1186/s12863-019-0802-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/15/2019] [Indexed: 01/09/2023] Open
Abstract
Background Corin is an important convertase involved in the natriuretic peptide system and may indirectly regulate blood pressure. Genetic factors relate to corin remain unclear. The purpose of the current study was to comprehensively examine the associations among CORIN SNPs, methylations, serum corin levels and hypertension. Results We genotyped 9 tag-SNPs in the CORIN gene and measured serum corin levels in 731 new-onset hypertensive cases and 731 age- and sex-matched controls. DNA methylations were tested in 43 individuals. Mendelian randomization was used to investigate the causal associations. Under additive models, we observed associations of rs2289433 (p.Cys13Tyr), rs6823184, rs10517195, rs2271037 and rs12509275 with serum corin levels after adjustment for covariates (P = 0.0399, 0.0238, 0.0016, 0.0148 and 0.0038, respectively). The tag-SNP rs6823184 and SNPs that are in strong linkage disequilibrium with it, i.e., rs10049713, rs6823698 and rs1866689, were associated with CORIN gene expression (P = 2.38 × 10− 24, 5.94 × 10− 27, 6.31 × 10− 27 and 6.30 × 10− 27, respectively). Neither SNPs nor corin levels was found to be associated with hypertension. SNP rs6823184, which is located in a DNase hypersensitivity cluster, a CpG island and transcription factor binding sites, was significantly associated with cg02955940 methylation levels (P = 1.54 × 10− 7). A putative causal association between cg02955940 methylation and corin levels was detected (P = 0.0011). Conclusion This study identified potentially functional CORIN SNPs that were associated with serum corin level in the Chinese Han population. The effect of CORIN SNPs on corin level may be mediated by DNA methylation.
Collapse
Affiliation(s)
- Huan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xingbo Mo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China.,Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Qiyu Qian
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Zhengyuan Zhou
- Changshu Center of Disease Control and Prevention, Suzhou, Jiangsu, People's Republic of China
| | - Zhengbao Zhu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xinfeng HuangFu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Tan Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Aili Wang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Zhirong Guo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Shufeng Lei
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China.,Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Yonghong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, Jiangsu, People's Republic of China. .,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
514
|
Damaging coding variants within kainate receptor channel genes are enriched in individuals with schizophrenia, autism and intellectual disabilities. Sci Rep 2019; 9:19215. [PMID: 31844109 PMCID: PMC6915710 DOI: 10.1038/s41598-019-55635-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/23/2019] [Indexed: 01/13/2023] Open
Abstract
Schizophrenia (Scz), autism spectrum disorder (ASD) and intellectual disability are common complex neurodevelopmental disorders. Kainate receptors (KARs) are ionotropic glutamate ion channels involved in synaptic plasticity which are modulated by auxiliary NETO proteins. Using UK10K exome sequencing data, we interrogated the coding regions of KAR and NETO genes in individuals with Scz, ASD or intellectual disability and population controls; performed follow-up genetic replication studies; and, conducted in silico and in vitro functional studies. We found an excess of Loss-of-Function and missense variants in individuals with Scz compared with control individuals (p = 1.8 × 10−10), and identified a significant burden of functional variants for Scz (p < 1.6 × 10−11) and ASD (p = 6.9 × 10−18). Single allele associations for 6 damaging missense variants were significantly replicated (p < 5.0 × 10−15) and confirmed GRIK3 S310A as a protective genetic factor. Functional studies demonstrated that three missense variants located within GluK2 and GluK4, GluK2 (K525E) and GluK4 (Y555N, L825W), affect agonist sensitivity and current decay rates. These findings establish that genetic variation in KAR receptor ion channels confers risk for schizophrenia, autism and intellectual disability and provide new genetic and pharmacogenetic biomarkers for neurodevelopmental disease.
Collapse
|
515
|
Zeng P, Wang T, Zheng J, Zhou X. Causal association of type 2 diabetes with amyotrophic lateral sclerosis: new evidence from Mendelian randomization using GWAS summary statistics. BMC Med 2019; 17:225. [PMID: 31796040 PMCID: PMC6892209 DOI: 10.1186/s12916-019-1448-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Associations between type 2 diabetes (T2D) and amyotrophic lateral sclerosis (ALS) were discovered in observational studies in both European and East Asian populations. However, whether such associations are causal remains largely unknown. METHODS We employed a two-sample Mendelian randomization approach to evaluate the causal relationship of T2D with the risk of ALS in both European and East Asian populations. Our analysis was implemented using summary statistics obtained from large-scale genome-wide association studies with ~660,000 individuals for T2D and ~81,000 individuals for ALS in the European population, and ~191,000 individuals for T2D and ~4100 individuals for ALS in the East Asian population. The causal relationship between T2D and ALS in both populations was estimated using the inverse-variance-weighted methods and was further validated through extensive complementary and sensitivity analyses. RESULTS Using multiple instruments that were strongly associated with T2D, a negative association between T2D and ALS was identified in the European population with the odds ratio (OR) estimated to be 0.93 (95% CI 0.88-0.99, p = 0.023), while a positive association between T2D and ALS was observed in the East Asian population with OR = 1.28 (95% CI 0.99-1.62, p = 0.058). These results were robust against instrument selection, various modeling misspecifications, and estimation biases, with the Egger regression and MR-PRESSO ruling out the possibility of horizontal pleiotropic effects of instruments. However, no causal association was found between T2D-related exposures (including glycemic traits) and ALS in the European population. CONCLUSION Our results provide new evidence supporting the causal neuroprotective role of T2D on ALS in the European population and provide empirically suggestive evidence of increasing risk of T2D on ALS in the East Asian population. Our results have an important implication on ALS pathology, paving ways for developing therapeutic strategies across multiple populations.
Collapse
Affiliation(s)
- Ping Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Ting Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA. .,Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
516
|
Li X, Wang H, Russell A, Cao W, Wang X, Ge S, Zheng Y, Guo Z, Hou H, Song M, Yu X, Wang Y, Hunter M, Roberts P, Lauc G, Wang W. Type 2 Diabetes Mellitus is Associated with the Immunoglobulin G N-Glycome through Putative Proinflammatory Mechanisms in an Australian Population. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 23:631-639. [PMID: 31526239 DOI: 10.1089/omi.2019.0075] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a common complex trait arising from interactions among multiple environmental, genomic, and postgenomic factors. We report here the first attempt to investigate the association between immunoglobulin G (IgG) N-glycan patterns, T2DM, and their clinical risk factors in an Australian population. N-glycosylation of proteins is one of the most frequently observed co- and post-translational modifications, reflecting, importantly, the real-time status of the interplay between the genomic and postgenomic factors. In a community-based case-control study, 849 participants (217 cases and 632 controls) were recruited from an urban community in Busselton, Western Australia. We applied the ultraperformance liquid chromatography method to analyze the composition of IgG N-glycans. We then conducted Spearman's correlation analyses to explore the association between glycan biomarker candidates and clinical risk factors. We performed area under the curve (AUC) analysis of the receiver operating characteristic curves by fivefold cross-validation for clinical risk factors, IgG glycans, and their combination. Two directly measured and four derived glycan peaks were significantly associated with T2DM, after correction for extensive clinical confounders and false discovery rate, thus suggesting that IgG N-glycan traits are highly correlated with T2DM clinical risk factors. Moreover, adding the IgG glycan profiles to fasting blood glucose in the logistic regression model increased the AUC from 0.799 to 0.859. The AUC for IgG glycans alone was 0.623 with a 95% confidence interval 0.580-0.666. In addition, our study provided new evidence of diversity in T2DM complex trait by IgG N-glycan stratification. Six IgG glycan traits were firmly associated with T2DM, which reflects an increased proinflammatory and biological aging status. In summary, our study reports novel associations between the IgG N-glycome and T2DM in an Australian population and the putative role of proinflammatory mechanisms. Furthermore, IgG N-glycomic alterations offer future prospects as inflammatory biomarker candidates for T2DM diagnosis, and monitoring of T2DM progression to cardiovascular disease or renal failure.
Collapse
Affiliation(s)
- Xingang Li
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Hao Wang
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Alyce Russell
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- School of Population and Global Health, University of Western Australia, Crawley, Australia
| | - Weijie Cao
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Xueqing Wang
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Siqi Ge
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yulu Zheng
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Zheng Guo
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Haifeng Hou
- School of Public Health, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
| | - Manshu Song
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Xinwei Yu
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- Tiantan Hospital, Capital Medical University, Beijing, China
| | - Youxin Wang
- Beijing Key Laboratory of Clinical Epidemiology, School of Public Health, Capital Medical University, Beijing, China
| | - Michael Hunter
- School of Population and Global Health, University of Western Australia, Crawley, Australia
- Busselton Health Study Centre, Busselton Population Medical Research Institute, Busselton, Australia
| | - Peter Roberts
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, BIOCentar, Zagreb, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Wei Wang
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- School of Public Health, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, China
- The First Affiliated Hospital, Shantou University Medical College, Shantou, China
| |
Collapse
|
517
|
Potential Impact of MicroRNA Gene Polymorphisms in the Pathogenesis of Diabetes and Atherosclerotic Cardiovascular Disease. J Pers Med 2019; 9:jpm9040051. [PMID: 31775219 PMCID: PMC6963792 DOI: 10.3390/jpm9040051] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are endogenous, small (18–23 nucleotides), non-coding RNA molecules. They regulate the posttranscriptional expression of their target genes. MiRNAs control vital physiological processes such as metabolism, development, differentiation, cell cycle and apoptosis. The control of the gene expression by miRNAs requires efficient binding between the miRNA and their target mRNAs. Genome-wide association studies (GWASs) have suggested the association of single-nucleotide polymorphisms (SNPs) with certain diseases in various populations. Gene polymorphisms of miRNA target sites have been implicated in diseases such as cancers, diabetes, cardiovascular and Parkinson’s disease. Likewise, gene polymorphisms of miRNAs have been reported to be associated with diseases. In this review, we discuss the SNPs in miRNA genes that have been associated with diabetes and atherosclerotic cardiovascular disease in different populations. We also discuss briefly the potential underlining mechanisms through which these SNPs increase the risk of developing these diseases.
Collapse
|
518
|
Molecular docking of polyoxometalates as potential α-glucosidase inhibitors. J Inorg Biochem 2019; 203:110914. [PMID: 31751818 DOI: 10.1016/j.jinorgbio.2019.110914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/30/2019] [Accepted: 11/08/2019] [Indexed: 01/16/2023]
Abstract
α-Glucosidase is an important target enzyme for the treatment of type 2 diabetes in humans. In our previous studies, it was found that polyoxometalates exhibited an effective inhibitory effect on the activity of α-glucosidase, while polyoxometalates have the characteristics of structural diversity and unique properties. Herein, we investigated the inhibition of two different series of polyoxometalates on α-glucosidases by enzyme kinetics and molecular docking. The results demonstrated that all of the studied compounds had a significant inhibitory ability on α-glucosidase as compared with the positive control acarbose. H8[P2Mo17Cr(OH2)O61] reversibly inhibited α-glucosidase in a competitive manner with IC50 of 115.50 ± 1.64 μM and KI value of 44.31 μM. All other compounds reversibly inhibited enzymatic activity in a mixed manner. H6PMo9V3O40 and H8[P2Mo17Cu(OH2)O61] were the best inhibitors in the Keggin and Dawson series, respectively, with IC50 of 9.63 ± 0.43 and 40.13 ± 0.61 μM, respectively. We conducted molecular docking study and found that the compound and α-glucosidase were mainly non-covalently interacting with hydrogen bonds and van der Waals forces. This result further confirmed the inhibition mechanism of enzyme kinetic experiments.
Collapse
|
519
|
Bains V, Kaur H, Badaruddoza. Association study of the single‐nucleotide polymorphisms −3971G/A and +276G/T in the adiponectin gene with type 2 diabetes in a North Indian Punjabi population. Ann Hum Genet 2019; 84:235-248. [DOI: 10.1111/ahg.12366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 07/16/2019] [Accepted: 10/28/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Veena Bains
- Department of Human Genetics Guru Nanak Dev University Amritsar Punjab India
| | - Harjit Kaur
- Department of Human Genetics Guru Nanak Dev University Amritsar Punjab India
| | - Badaruddoza
- Department of Human Genetics Guru Nanak Dev University Amritsar Punjab India
| |
Collapse
|
520
|
Beijer K, Nowak C, Sundström J, Ärnlöv J, Fall T, Lind L. In search of causal pathways in diabetes: a study using proteomics and genotyping data from a cross-sectional study. Diabetologia 2019; 62:1998-2006. [PMID: 31446444 PMCID: PMC6805963 DOI: 10.1007/s00125-019-4960-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 06/06/2019] [Indexed: 12/12/2022]
Abstract
AIMS/HYPOTHESIS The pathogenesis of type 2 diabetes is not fully understood. We investigated whether circulating levels of preselected proteins were associated with the outcome 'diabetes' and whether these associations were causal. METHODS In 2467 individuals of the population-based, cross-sectional EpiHealth study (45-75 years, 50% women), 249 plasma proteins were analysed by the proximity extension assay technique. DNA was genotyped using the Illumina HumanCoreExome-12 v1.0 BeadChip. Diabetes was defined as taking glucose-lowering treatment or having a fasting plasma glucose of ≥7.0 mmol/l. The associations between proteins and diabetes were assessed using logistic regression. To investigate causal relationships between proteins and diabetes, a bidirectional two-sample Mendelian randomisation was performed based on large, genome-wide association studies belonging to the DIAGRAM and MAGIC consortia, and a genome-wide association study in the EpiHealth study. RESULTS Twenty-six proteins were positively associated with diabetes, including cathepsin D, retinal dehydrogenase 1, α-L-iduronidase, hydroxyacid oxidase 1 and galectin-4 (top five findings). Three proteins, lipoprotein lipase, IGF-binding protein 2 and paraoxonase 3 (PON-3), were inversely associated with diabetes. Fourteen of the proteins are novel discoveries. The Mendelian randomisation study did not disclose any significant causal effects between the proteins and diabetes in either direction that were consistent with the relationships found between the protein levels and diabetes. CONCLUSIONS/INTERPRETATION The 29 proteins associated with diabetes are involved in several physiological pathways, but given the power of the study no causal link was identified for those proteins tested in Mendelian randomisation. Therefore, the identified proteins are likely to be biomarkers for type 2 diabetes, rather than representing causal pathways.
Collapse
Affiliation(s)
- Kristina Beijer
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden.
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society, Division of Family Medicine and Primary Care, Karolinska Institute, Stockholm, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Division of Family Medicine and Primary Care, Karolinska Institute, Stockholm, Sweden
- School of Health and Social Sciences, Dalarna University, Falun, Sweden
| | - Tove Fall
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, UCR, Dag Hammarskjölds väg 38, SE-751 83, Uppsala, Sweden
| |
Collapse
|
521
|
Thompson DJ, Genovese G, Halvardson J, Ulirsch JC, Wright DJ, Terao C, Davidsson OB, Day FR, Sulem P, Jiang Y, Danielsson M, Davies H, Dennis J, Dunlop MG, Easton DF, Fisher VA, Zink F, Houlston RS, Ingelsson M, Kar S, Kerrison ND, Kinnersley B, Kristjansson RP, Law PJ, Li R, Loveday C, Mattisson J, McCarroll SA, Murakami Y, Murray A, Olszewski P, Rychlicka-Buniowska E, Scott RA, Thorsteinsdottir U, Tomlinson I, Moghadam BT, Turnbull C, Wareham NJ, Gudbjartsson DF, Kamatani Y, Hoffmann ER, Jackson SP, Stefansson K, Auton A, Ong KK, Machiela MJ, Loh PR, Dumanski JP, Chanock SJ, Forsberg LA, Perry JRB. Genetic predisposition to mosaic Y chromosome loss in blood. Nature 2019; 575:652-657. [PMID: 31748747 PMCID: PMC6887549 DOI: 10.1038/s41586-019-1765-3] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022]
Abstract
Mosaic loss of chromosome Y (LOY) in circulating white blood cells is the most common form of clonal mosaicism1-5, yet our knowledge of the causes and consequences of this is limited. Here, using a computational approach, we estimate that 20% of the male population represented in the UK Biobank study (n = 205,011) has detectable LOY. We identify 156 autosomal genetic determinants of LOY, which we replicate in 757,114 men of European and Japanese ancestry. These loci highlight genes that are involved in cell-cycle regulation and cancer susceptibility, as well as somatic drivers of tumour growth and targets of cancer therapy. We demonstrate that genetic susceptibility to LOY is associated with non-haematological effects on health in both men and women, which supports the hypothesis that clonal haematopoiesis is a biomarker of genomic instability in other tissues. Single-cell RNA sequencing identifies dysregulated expression of autosomal genes in leukocytes with LOY and provides insights into why clonal expansion of these cells may occur. Collectively, these data highlight the value of studying clonal mosaicism to uncover fundamental mechanisms that underlie cancer and other ageing-related diseases.
Collapse
Affiliation(s)
- Deborah J Thompson
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Giulio Genovese
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonatan Halvardson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jacob C Ulirsch
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Daniel J Wright
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Open Targets Core Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Chikashi Terao
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | | | - Felix R Day
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | - Marcus Danielsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Hanna Davies
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Malcolm G Dunlop
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit and CRUK Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Victoria A Fisher
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Martin Ingelsson
- Geriatrics Research Group, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Siddhartha Kar
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Nicola D Kerrison
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ben Kinnersley
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | | | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Rong Li
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chey Loveday
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Jonas Mattisson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Steven A McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Anna Murray
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Pawel Olszewski
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Edyta Rychlicka-Buniowska
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Robert A Scott
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Ian Tomlinson
- Cancer Genetics and Evolution Laboratory, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Behrooz Torabi Moghadam
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Clare Turnbull
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- William Harvey Research Institute, Queen Mary University, London, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Daniel F Gudbjartsson
- deCODE Genetics, Amgen, Reykjavík, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavík, Iceland
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Eva R Hoffmann
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steve P Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Wellcome Trust and Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Kari Stefansson
- deCODE Genetics, Amgen, Reykjavík, Iceland
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | - Ken K Ong
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Po-Ru Loh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jan P Dumanski
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Lars A Forsberg
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Beijer Laboratory of Genome Research, Uppsala University, Uppsala, Sweden
| | - John R B Perry
- MRC Epidemiology Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
522
|
Tremblay J, Hamet P. Environmental and genetic contributions to diabetes. Metabolism 2019; 100S:153952. [PMID: 31610851 DOI: 10.1016/j.metabol.2019.153952] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 01/18/2023]
Abstract
Diabetes mellitus (DM) is a heterogeneous group of disorders characterized by persistent hyperglycemia. Its two most common forms are type 1 diabetes (T1D) and type 2 diabetes (T2D), for which genetic and environmental risk factors act in synergy. Because it occurs in children and involves infectious, autoimmune or toxic destruction of the insulin-secreting pancreatic beta-cells, type 1 diabetes has been called juvenile or insulin-deficient diabetes. In type 2, patients can still secrete some insulin but its effectiveness may be attenuated by 'insulin resistance.' There is also a group of rare forms of diabetes in the young which are inherited as monogenetic diseases. Whether one calls the underlying process 'genes vs. environment' or 'nature vs nurture', diabetes occurs at the interface of the two domains. Together with our genetic background we are born tabula rasa-a blank slate upon which the story of life, with all its environmental inputs will be written. There is one proviso: the influence of epigenetic inheritance must also be considered. Thus, in the creation of databases that include "big data" originating from genomic as well as exposome (defined as: the totality of environmental exposure from conception to death), a broad perspective is crucial as these factors act in concert in such chronic illnesses as diabetes that, for example, are likely to require adoption of an appropriate lifestyle change. Also, it is becoming increasingly evident that epigenetic factors can modulate the interplay between genes and environment. Consequently, throughout the life of an individual nature and nurture interact in a complex manner in the development of diabetes. This review addresses the question of the contribution of gene and environment and their interactions in the development of diabetes.
Collapse
Affiliation(s)
- Johanne Tremblay
- CRCHUM Research Center, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Pavel Hamet
- CRCHUM Research Center, Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
523
|
Abstract
The two-phase design is a cost-effective sampling strategy to evaluate the effects of covariates on an outcome when certain covariates are too expensive to be measured on all study subjects. Under such a design, the outcome and inexpensive covariates are measured on all subjects in the first phase and the first-phase information is used to select subjects for measurements of expensive covariates in the second phase. Previous research on two-phase studies has focused largely on the inference procedures rather than the design aspects. We investigate the design efficiency of the two-phase study, as measured by the semiparametric efficiency bound for estimating the regression coefficients of expensive covariates. We consider general two-phase studies, where the outcome variable can be continuous, discrete, or censored, and the second-phase sampling can depend on the first-phase data in any manner. We develop optimal or approximately optimal two-phase designs, which can be substantially more efficient than the existing designs. We demonstrate the improvements of the new designs over the existing ones through extensive simulation studies and two large medical studies.
Collapse
Affiliation(s)
- Ran Tao
- Department of Biostatistics and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232.,Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599
| | - Donglin Zeng
- Department of Biostatistics and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232.,Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599
| | - Dan-Yu Lin
- Department of Biostatistics and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232.,Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
524
|
Raulerson CK, Ko A, Kidd JC, Currin KW, Brotman SM, Cannon ME, Wu Y, Spracklen CN, Jackson AU, Stringham HM, Welch RP, Fuchsberger C, Locke AE, Narisu N, Lusis AJ, Civelek M, Furey TS, Kuusisto J, Collins FS, Boehnke M, Scott LJ, Lin DY, Love MI, Laakso M, Pajukanta P, Mohlke KL. Adipose Tissue Gene Expression Associations Reveal Hundreds of Candidate Genes for Cardiometabolic Traits. Am J Hum Genet 2019; 105:773-787. [PMID: 31564431 PMCID: PMC6817527 DOI: 10.1016/j.ajhg.2019.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with cardiometabolic traits including type 2 diabetes (T2D), lipid levels, body fat distribution, and adiposity, although most causal genes remain unknown. We used subcutaneous adipose tissue RNA-seq data from 434 Finnish men from the METSIM study to identify 9,687 primary and 2,785 secondary cis-expression quantitative trait loci (eQTL; <1 Mb from TSS, FDR < 1%). Compared to primary eQTL signals, secondary eQTL signals were located further from transcription start sites, had smaller effect sizes, and were less enriched in adipose tissue regulatory elements compared to primary signals. Among 2,843 cardiometabolic GWAS signals, 262 colocalized by LD and conditional analysis with 318 transcripts as primary and conditionally distinct secondary cis-eQTLs, including some across ancestries. Of cardiometabolic traits examined for adipose tissue eQTL colocalizations, waist-hip ratio (WHR) and circulating lipid traits had the highest percentage of colocalized eQTLs (15% and 14%, respectively). Among alleles associated with increased cardiometabolic GWAS risk, approximately half (53%) were associated with decreased gene expression level. Mediation analyses of colocalized genes and cardiometabolic traits within the 434 individuals provided further evidence that gene expression influences variant-trait associations. These results identify hundreds of candidate genes that may act in adipose tissue to influence cardiometabolic traits.
Collapse
Affiliation(s)
- Chelsea K Raulerson
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Arthur Ko
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - John C Kidd
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kevin W Currin
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah M Brotman
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Maren E Cannon
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryan P Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christian Fuchsberger
- Center for Biomedicine, European Academy of Bolzano/Bozen, University of Lübeck, Bolzano/Bozen 39100, Italy
| | - Adam E Locke
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Narisu Narisu
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aldons J Lusis
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Terrence S Furey
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio 70210, Finland
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dan-Yu Lin
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Kuopio University Hospital, University of Eastern Finland, Kuopio 70210, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
525
|
Serotonin is elevated in risk-genotype carriers of TCF7L2 - rs7903146. Sci Rep 2019; 9:12863. [PMID: 31492908 PMCID: PMC6731216 DOI: 10.1038/s41598-019-49347-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/12/2019] [Indexed: 01/21/2023] Open
Abstract
The transcription factor 7-like 2 (TCF7L2) polymorphism rs7903146 is known to be tightly associated with an elevated risk for type 2 diabetes, whereas the molecular mechanisms remain elusive. We evaluated the metabolic profile of a total of 394 patients' serum samples with respect to their rs7903146 genotype using targeted metabolomics in a discovery (n = 154) and a validation (n = 240) study. We have identified serotonin as the top metabolite being increased in carriers of the risk allele. Serotonin was significantly associated with the rs7903146 genotype after full adjustment including type 2 diabetes and further top ranked metabolites. Given the role of peripheral serotonin in metabolic homeostasis and type 2 diabetes, this finding provides a first hint that the well-known impact of the TCF7L2 polymorphism on type 2 diabetes risk may involve a serotonin-dependent pathway.
Collapse
|
526
|
Anderson K, Cañadas-Garre M, Chambers R, Maxwell AP, McKnight AJ. The Challenges of Chromosome Y Analysis and the Implications for Chronic Kidney Disease. Front Genet 2019; 10:781. [PMID: 31552093 PMCID: PMC6737325 DOI: 10.3389/fgene.2019.00781] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
The role of chromosome Y in chronic kidney disease (CKD) remains unknown, as chromosome Y is typically excluded from genetic analysis in CKD. The complex, sex-specific presentation of CKD could be influenced by chromosome Y genetic variation, but there is limited published research available to confirm or reject this hypothesis. Although traditionally thought to be associated with male-specific disease, evidence linking chromosome Y genetic variation to common complex disorders highlights a potential gap in CKD research. Chromosome Y variation has been associated with cardiovascular disease, a condition closely linked to CKD and one with a very similar sexual dimorphism. Relatively few sources of genetic variation in chromosome Y have been examined in CKD. The association between chromosome Y aneuploidy and CKD has never been explored comprehensively, while analyses of microdeletions, copy number variation, and single-nucleotide polymorphisms in CKD have been largely limited to the autosomes or chromosome X. In many studies, it is unclear whether the analyses excluded chromosome Y or simply did not report negative results. Lack of imputation, poor cross-study comparability, and requirement for separate or additional analyses in comparison with autosomal chromosomes means that chromosome Y is under-investigated in the context of CKD. Limitations in genotyping arrays could be overcome through use of whole-chromosome sequencing of chromosome Y that may allow analysis of many different types of genetic variation across the chromosome to determine if chromosome Y genetic variation is associated with CKD.
Collapse
Affiliation(s)
- Kerry Anderson
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, c/o Regional Genetics Centre, Belfast City Hospital, Belfast, United Kingdom
| | - Marisa Cañadas-Garre
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, c/o Regional Genetics Centre, Belfast City Hospital, Belfast, United Kingdom
| | - Robyn Chambers
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, c/o Regional Genetics Centre, Belfast City Hospital, Belfast, United Kingdom
| | - Alexander Peter Maxwell
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, c/o Regional Genetics Centre, Belfast City Hospital, Belfast, United Kingdom.,Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom
| | - Amy Jayne McKnight
- Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University of Belfast, c/o Regional Genetics Centre, Belfast City Hospital, Belfast, United Kingdom
| |
Collapse
|
527
|
Huang Z, Tan R, Meng L, Yang H, Liang X, Qin Y, Luo Z. The Niemann-Pick C1-like 1 rs2073547 polymorphism is associated with type 2 diabetes mellitus in a Chinese population. J Int Med Res 2019; 47:4260-4271. [PMID: 31311377 PMCID: PMC6753537 DOI: 10.1177/0300060519862099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective To explore the association between Niemann–Pick C1-like 1 gene ( NPC1L1) single nucleotide polymorphisms and type 2 diabetes mellitus (T2DM) in a Chinese population. Methods Using propensity score matching, 490 T2DM patients and 490 matched controls were recruited from 13 communities in Guangxi, China. NPC1L1 rs217386 and rs2073547 genotyping was performed using a MassARRAY system. Results The rs2073547 genotype distribution differed significantly among patient groups. Low-density lipoprotein cholesterol levels were similar among different rs2073547 genotypes and alleles. The rs2073547 AG genotype was significantly more prevalent in patients with T2DM. After adjusting for risk or protective factors for diabetes, AG and GG+AG genotypes of rs2073547 were associated with significantly increased risks of T2DM. Compared with the AA genotype, the AG genotype was associated with a significantly higher risk of T2DM in participants with gamma-glutamyl transpeptidase (GGT) <45 U/L, systolic blood pressure (SBP) ≥140 mmHg, or triglyceride <1.70 mmol/L. In participants with GGT <45 U/L or SBP ≥140 mmHg, the GG+AG genotype was associated with a significantly higher T2DM risk versus the AA genotype. Conclusions The rs2073547 polymorphism of NPC1L1 may be related to T2DM susceptibility in the Chinese population.
Collapse
Affiliation(s)
- Zhenxing Huang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Ruyin Tan
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Liheng Meng
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Haiyan Yang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xinghuan Liang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yingfen Qin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zuojie Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
528
|
Brzozowska MM, Havula E, Allen RB, Cox MP. Genetics, adaptation to environmental changes and archaic admixture in the pathogenesis of diabetes mellitus in Indigenous Australians. Rev Endocr Metab Disord 2019; 20:321-332. [PMID: 31278514 DOI: 10.1007/s11154-019-09505-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Indigenous Australians are particularly affected by type 2 diabetes mellitus (T2D) due to both their genetic susceptibility and a range of environmental and lifestyle risk factors. Recent genetic studies link predisposition to some diseases, including T2D, to alleles acquired from archaic hominins, such as Neanderthals and Denisovans, which persist in the genomes of modern humans today. Indo-Pacific human populations, including Indigenous Australians, remain extremely underrepresented in genomic research with a paucity of data examining the impact of Denisovan or Neanderthal lineages on human phenotypes in Oceania. The few genetic studies undertaken emphasize the uniqueness and antiquity of Indigenous Australian genomes, with possibly the largest proportion of Denisovan ancestry of any population in the world. In this review, we focus on the potential contributions of ancient genes/pathways to modern human phenotypes, while also highlighting the evolutionary roles of genetic adaptation to dietary and environmental changes associated with an adopted Western lifestyle. We discuss the role of genetic and epigenetic factors in the pathogenesis of T2D in understudied Indigenous Australians, including the potential impact of archaic gene lineages on this disease. Finally, we propose that greater understanding of the underlying genetic predisposition may contribute to the clinical efficacy of diabetes management in Indigenous Australians. We suggest that improved identification of T2D risk variants in Oceania is needed. Such studies promise to clarify how genetic and phenotypic differences vary between populations and, crucially, provide novel targets for personalised medical therapies in currently marginalized groups.
Collapse
Affiliation(s)
- Malgorzata Monika Brzozowska
- Endocrinology Department, Sutherland Hospital, Sydney, New South Wales, Australia.
- St George & Sutherland Hospital Clinical School, University of New South Wales, Sydney, Australia.
| | - Essi Havula
- School of Life and Environmental Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Richard Benjamin Allen
- The Palaeogenomics and Bio-Archaeology Research Network, Research Laboratory for Archaeology and History of Art, University of Oxford, Oxford, UK
| | - Murray P Cox
- Statistics and Bioinformatics Group, School of Fundamental Sciences, Massey University, Palmerston North, 4410, New Zealand
| |
Collapse
|
529
|
Del Prato S. Heterogeneity of diabetes: heralding the era of precision medicine. Lancet Diabetes Endocrinol 2019; 7:659-661. [PMID: 31345775 DOI: 10.1016/s2213-8587(19)30218-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Stefano Del Prato
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases and Diabetes, Nuovo Ospedale Santa Chiara, University of Pisa, Pisa 56124, Italy.
| |
Collapse
|
530
|
Zhou Z, Zhu Y, Liu Y, Yin Y. Comprehensive transcriptomic analysis indicates brain regional specific alterations in type 2 diabetes. Aging (Albany NY) 2019; 11:6398-6421. [PMID: 31449493 PMCID: PMC6738403 DOI: 10.18632/aging.102196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/10/2019] [Indexed: 04/07/2023]
Abstract
Type 2 diabetes (T2D) can result in a number of comorbidities involving various organs including heart, eye, kidney and even the brain. However, little is known about the molecular basis of T2D associated brain disorders. In this study, we performed a comprehensive transcriptomic analysis in a total of 304 T2D samples and 608 matched control samples from thirteen distinct brain regions. We observed prominent difference among transcriptomic profiles of diverse brain regions in T2D. The most striking change was found in caudate with thousands of T2D-associated genes identified, followed by hippocampus, while nearly no transcriptomic change was observed in other brain regions. Functional analysis of T2D-associated genes revealed impaired synaptic functions and an association with neurodegenerative diseases. Co-expression analysis of caudate transcriptomic profiles unveiled a core regional specific module that was disorganized in T2D. Sub-modules consisting of regional markers were enriched in T2D risk single nucleotide polymorphisms (SNPs) and implied a causal link with T2D. Hub genes of this module include NSF and ADD2, the former of which has been associated with T2D and neurogenerative diseases. Thus, our work provides useful information for further studies in T2D associated brain disorders.
Collapse
Affiliation(s)
- Zhe Zhou
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yizhang Zhu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yang Liu
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
531
|
Ramezankhani A, Guity K, Azizi F, Hadaegh F. Sex differences in the association between spousal metabolic risk factors with incidence of type 2 diabetes: a longitudinal study of the Iranian population. Biol Sex Differ 2019; 10:41. [PMID: 31439024 PMCID: PMC6704543 DOI: 10.1186/s13293-019-0255-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 08/12/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND We investigated whether metabolic risk factors in one spouse were associated with an excessive risk of type 2 diabetes in the other. METHODS The study cohort (1999-2018) included 1833 men and 1952 women, aged ≥ 20 years with information on both their own and their spouse's diabetes status and metabolic risk factors including body mass index (BMI), waist circumference, systolic and diastolic blood pressure, triglyceride to high-density lipoprotein cholesterol ratio, and type 2 diabetes. The associations between spousal metabolic risk factors and type 2 diabetes were estimated using Cox regression models adjusted for the three nested sets of covariates. RESULTS We found 714 (360 men and 354 women) incident cases of type 2 diabetes, after more than 15 years of follow-up. Among women, having a husband with diabetes was associated with a 38% (hazard ratio (HR) 1.38; 95% confidence interval (CI) 1.03, 1. 84) increased risk of type 2 diabetes, adjusted for age, socioeconomic status, individual's own value of the respective spousal exposure variable, family history of diabetes, and physical activity level. After further adjustment for the woman's own BMI level, the husband's diabetes was associated with 23% (HR 1.23; 0.92, 1.64) higher risk of type 2 diabetes in wives, values which did not reach statistical significance. No significant associations were found between spousal metabolic risk factors and incidence of type 2 diabetes among index men. CONCLUSION We found a sex-specific effect of spousal diabetes on the risk of type 2 diabetes. Having a husband with diabetes increased an individual's risk of type 2 diabetes. Our results might contribute to the early detection of individuals at high risk of developing type 2 diabetes, particularly, in women adversely affected by their partner's diabetes.
Collapse
Affiliation(s)
- Azra Ramezankhani
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Guity
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
532
|
De Silva NMG, Borges MC, Hingorani AD, Engmann J, Shah T, Zhang X, Luan J, Langenberg C, Wong A, Kuh D, Chambers JC, Zhang W, Jarvelin MR, Sebert S, Auvinen J, Gaunt TR, Lawlor DA. Liver Function and Risk of Type 2 Diabetes: Bidirectional Mendelian Randomization Study. Diabetes 2019; 68:1681-1691. [PMID: 31088856 PMCID: PMC7011195 DOI: 10.2337/db18-1048] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/05/2019] [Indexed: 12/15/2022]
Abstract
Liver dysfunction and type 2 diabetes (T2D) are consistently associated. However, it is currently unknown whether liver dysfunction contributes to, results from, or is merely correlated with T2D due to confounding. We used Mendelian randomization to investigate the presence and direction of any causal relation between liver function and T2D risk including up to 64,094 T2D case and 607,012 control subjects. Several biomarkers were used as proxies of liver function (i.e., alanine aminotransferase [ALT], aspartate aminotransferase [AST], alkaline phosphatase [ALP], and γ-glutamyl transferase [GGT]). Genetic variants strongly associated with each liver function marker were used to investigate the effect of liver function on T2D risk. In addition, genetic variants strongly associated with T2D risk and with fasting insulin were used to investigate the effect of predisposition to T2D and insulin resistance, respectively, on liver function. Genetically predicted higher circulating ALT and AST were related to increased risk of T2D. There was a modest negative association of genetically predicted ALP with T2D risk and no evidence of association between GGT and T2D risk. Genetic predisposition to higher fasting insulin, but not to T2D, was related to increased circulating ALT. Since circulating ALT and AST are markers of nonalcoholic fatty liver disease (NAFLD), these findings provide some support for insulin resistance resulting in NAFLD, which in turn increases T2D risk.
Collapse
Affiliation(s)
- N Maneka G De Silva
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, U.K
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College London, London, U.K
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, U.K
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Aroon D Hingorani
- UCL Institute of Cardiovascular Science, Research Department of Population Science and Experimental Medicine, Centre for Translational Genomics, University College London, London, U.K
- Farr Institute, University College London, London, U.K
| | - Jorgen Engmann
- UCL Institute of Cardiovascular Science, Research Department of Population Science and Experimental Medicine, Centre for Translational Genomics, University College London, London, U.K
| | - Tina Shah
- UCL Institute of Cardiovascular Science, Research Department of Population Science and Experimental Medicine, Centre for Translational Genomics, University College London, London, U.K
| | - Xiaoshuai Zhang
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Binzhou Medical University, Yantai, Shandong, China
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, Shandong, China
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | | | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, U.K
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, U.K
| | - John C Chambers
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College London, London, U.K
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Epidemiology and Biostatistics, Imperial College London, London, U.K
- Department of Cardiology, Ealing Hospital, Middlesex, U.K
- Imperial College Healthcare NHS Trust, Imperial College London, London, U.K
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, U.K
- Department of Cardiology, Ealing Hospital, Middlesex, U.K
| | - Marjo-Ritta Jarvelin
- Department of Epidemiology and Biostatistics, Imperial College London, London, U.K
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
- Biocenter Oulu, Oulu, Finland
| | - Sylvain Sebert
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - Juha Auvinen
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
- Oulunkaari Health Center, Ii, Finland
| | | | - Tom R Gaunt
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, U.K
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, U.K.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| |
Collapse
|
533
|
Nasykhova YA, Barbitoff YA, Serebryakova EA, Katserov DS, Glotov AS. Recent advances and perspectives in next generation sequencing application to the genetic research of type 2 diabetes. World J Diabetes 2019; 10:376-395. [PMID: 31363385 PMCID: PMC6656706 DOI: 10.4239/wjd.v10.i7.376] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/23/2019] [Accepted: 06/11/2019] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes (T2D) mellitus is a common complex disease that currently affects more than 400 million people worldwide and has become a global health problem. High-throughput sequencing technologies such as whole-genome and whole-exome sequencing approaches have provided numerous new insights into the molecular bases of T2D. Recent advances in the application of sequencing technologies to T2D research include, but are not limited to: (1) Fine mapping of causal rare and common genetic variants; (2) Identification of confident gene-level associations; (3) Identification of novel candidate genes by specific scoring approaches; (4) Interrogation of disease-relevant genes and pathways by transcriptional profiling and epigenome mapping techniques; and (5) Investigation of microbial community alterations in patients with T2D. In this work we review these advances in application of next-generation sequencing methods for elucidation of T2D pathogenesis, as well as progress and challenges in implementation of this new knowledge about T2D genetics in diagnosis, prevention, and treatment of the disease.
Collapse
Affiliation(s)
- Yulia A Nasykhova
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
| | - Yury A Barbitoff
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Bioinformatics Institute, St. Petersburg 194021, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Elena A Serebryakova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
- Department of Genetics, City Hospital No. 40, St. Petersburg 197706, Russia
| | - Dmitry S Katserov
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad 236016, Russia
| | - Andrey S Glotov
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg 199034, Russia
- Department of Genetics, City Hospital No. 40, St. Petersburg 197706, Russia
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad 236016, Russia
| |
Collapse
|
534
|
Verma S, Rizvi S, Abbas M, Raza T, Mahdi F. Personalized medicine- future of diagnosis and management of T2DM. Diabetes Metab Syndr 2019; 13:2425-2430. [PMID: 31405654 DOI: 10.1016/j.dsx.2019.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/12/2019] [Indexed: 11/24/2022]
Affiliation(s)
- Sushma Verma
- Department of Personalized and Molecular Medicine, Era University, Lucknow, 226003, Uttar Pradesh, India.
| | - Saliha Rizvi
- Department of Personalized and Molecular Medicine, Era University, Lucknow, 226003, Uttar Pradesh, India.
| | - Mohd Abbas
- Department of Microbiology, Era University, Lucknow, 226003, Uttar Pradesh, India.
| | - Tasleem Raza
- Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, Uttar Pradesh, India.
| | - Farzana Mahdi
- Department of Personalized and Molecular Medicine, Era University, Lucknow, 226003, Uttar Pradesh, India.
| |
Collapse
|
535
|
Samaha G, Beatty J, Wade CM, Haase B. The Burmese cat as a genetic model of type 2 diabetes in humans. Anim Genet 2019; 50:319-325. [PMID: 31179570 DOI: 10.1111/age.12799] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2019] [Indexed: 12/16/2022]
Abstract
The recent extension of genetic tools to the domestic cat, together with the serendipitous consequences of selective breeding, have been essential to the study of the genetic diseases that affect them. Cats are increasingly presented for veterinary surveillance and share many of human's heritable diseases, allowing them to serve as natural models of these conditions. Feline diabetes mellitus is a common condition in domestic cats that bears close pathological and clinical resemblance to type 2 diabetes in humans, including pancreatic β-cell dysfunction and peripheral insulin resistance. In Australia, New Zealand and Europe, diabetes mellitus is almost four times more common in cats of the Burmese breed than in other breeds. This geographically based breed predisposition parallels familial and population clustering of type 2 diabetes in humans. As a genetically isolated population, the Australian Burmese breed provides a spontaneous, naturally occurring genetic model of type 2 diabetes. Genetically isolated populations typically exhibit extended linkage disequilibrium and increased opportunity for deleterious variants to reach high frequencies over many generations due to genetic drift. Studying complex diseases in such populations allows for tighter control of confounding factors including environmental heterogeneity, allelic frequencies and population stratification. The homogeneous genetic background of Australian Burmese cats may provide a unique opportunity to either refine genetic signals previously associated with type 2 diabetes or identify new risk factors for this disease.
Collapse
Affiliation(s)
- G Samaha
- Sydney School of Veterinary Science, University of Sydney, Sydney, NSW, 2006, Australia
| | - J Beatty
- Sydney School of Veterinary Science, Valentine Charlton Cat Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - C M Wade
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - B Haase
- Sydney School of Veterinary Science, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
536
|
Missing heritability of complex diseases: case solved? Hum Genet 2019; 139:103-113. [DOI: 10.1007/s00439-019-02034-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/28/2019] [Indexed: 10/26/2022]
|
537
|
Gupta MB, Jansson T. Novel roles of mechanistic target of rapamycin signaling in regulating fetal growth†. Biol Reprod 2019; 100:872-884. [PMID: 30476008 PMCID: PMC6698747 DOI: 10.1093/biolre/ioy249] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/08/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) signaling functions as a central regulator of cellular metabolism, growth, and survival in response to hormones, growth factors, nutrients, energy, and stress signals. Mechanistic TOR is therefore critical for the growth of most fetal organs, and global mTOR deletion is embryonic lethal. This review discusses emerging evidence suggesting that mTOR signaling also has a role as a critical hub in the overall homeostatic control of fetal growth, adjusting the fetal growth trajectory according to the ability of the maternal supply line to support fetal growth. In the fetus, liver mTOR governs the secretion and phosphorylation of insulin-like growth factor binding protein 1 (IGFBP-1) thereby controlling the bioavailability of insulin-like growth factors (IGF-I and IGF-II), which function as important growth hormones during fetal life. In the placenta, mTOR responds to a large number of growth-related signals, including amino acids, glucose, oxygen, folate, and growth factors, to regulate trophoblast mitochondrial respiration, nutrient transport, and protein synthesis, thereby influencing fetal growth. In the maternal compartment, mTOR is an integral part of a decidual nutrient sensor which links oxygen and nutrient availability to the phosphorylation of IGFBP-1 with preferential effects on the bioavailability of IGF-I in the maternal-fetal interface and in the maternal circulation. These new roles of mTOR signaling in the regulation fetal growth will help us better understand the molecular underpinnings of abnormal fetal growth, such as intrauterine growth restriction and fetal overgrowth, and may represent novel avenues for diagnostics and intervention in important pregnancy complications.
Collapse
Affiliation(s)
- Madhulika B Gupta
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
| | - Thomas Jansson
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, University of Colorado | Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
538
|
Barroso I, McCarthy MI. The Genetic Basis of Metabolic Disease. Cell 2019; 177:146-161. [PMID: 30901536 PMCID: PMC6432945 DOI: 10.1016/j.cell.2019.02.024] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/11/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023]
Abstract
Recent developments in genetics and genomics are providing a detailed and systematic characterization of the genetic underpinnings of common metabolic diseases and traits, highlighting the inherent complexity within systems for homeostatic control and the many ways in which that control can fail. The genetic architecture underlying these common metabolic phenotypes is complex, with each trait influenced by hundreds of loci spanning a range of allele frequencies and effect sizes. Here, we review the growing appreciation of this complexity and how this has fostered the implementation of genome-scale approaches that deliver robust mechanistic inference and unveil new strategies for translational exploitation.
Collapse
Affiliation(s)
- Inês Barroso
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK.
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; Oxford Centre for Diabetes, Endocrinology, and Metabolism, University of Oxford, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Old Road, Headington, Oxford OX3 7LJ, UK
| |
Collapse
|
539
|
Suzuki K, Akiyama M, Ishigaki K, Kanai M, Hosoe J, Shojima N, Hozawa A, Kadota A, Kuriki K, Naito M, Tanno K, Ishigaki Y, Hirata M, Matsuda K, Iwata N, Ikeda M, Sawada N, Yamaji T, Iwasaki M, Ikegawa S, Maeda S, Murakami Y, Wakai K, Tsugane S, Sasaki M, Yamamoto M, Okada Y, Kubo M, Kamatani Y, Horikoshi M, Yamauchi T, Kadowaki T. Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019. [DOI: 10.1038/s41588-018-0332-4 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
540
|
Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019. [DOI: 10.1038/s41588-018-0332-4 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
541
|
Suzuki K, Akiyama M, Ishigaki K, Kanai M, Hosoe J, Shojima N, Hozawa A, Kadota A, Kuriki K, Naito M, Tanno K, Ishigaki Y, Hirata M, Matsuda K, Iwata N, Ikeda M, Sawada N, Yamaji T, Iwasaki M, Ikegawa S, Maeda S, Murakami Y, Wakai K, Tsugane S, Sasaki M, Yamamoto M, Okada Y, Kubo M, Kamatani Y, Horikoshi M, Yamauchi T, Kadowaki T. Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019. [DOI: 10.1038/s41588-018-0332-4 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
542
|
Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019. [DOI: 10.1038/s41588-018-0332-4 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
543
|
Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019. [DOI: 10.1038/s41588-018-0332-4 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
544
|
Suzuki K, Akiyama M, Ishigaki K, Kanai M, Hosoe J, Shojima N, Hozawa A, Kadota A, Kuriki K, Naito M, Tanno K, Ishigaki Y, Hirata M, Matsuda K, Iwata N, Ikeda M, Sawada N, Yamaji T, Iwasaki M, Ikegawa S, Maeda S, Murakami Y, Wakai K, Tsugane S, Sasaki M, Yamamoto M, Okada Y, Kubo M, Kamatani Y, Horikoshi M, Yamauchi T, Kadowaki T. Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019. [DOI: 10.1038/s41588-018-0332-4 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
545
|
Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019. [DOI: 10.1038/s41588-018-0332-4 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
546
|
Mineral Nutrition and the Risk of Chronic Diseases: A Mendelian Randomization Study. Nutrients 2019; 11:nu11020378. [PMID: 30759836 PMCID: PMC6412267 DOI: 10.3390/nu11020378] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
We applied Mendelian randomization analyses to investigate the potential causality between blood minerals (calcium, magnesium, iron, copper, and zinc) and osteoporosis (OP), gout, rheumatoid arthritis (RA), type 2 diabetes (T2D), Alzheimer’s disease (AD), bipolar disorder (BD), schizophrenia, Parkinson’s disease and major depressive disorder. Single nucleotide polymorphisms (SNPs) that are independent (r2 < 0.01) and are strongly related to minerals (p < 5 × 10−8) are selected as instrumental variables. Each standard deviation increase in magnesium (0.16 mmol/L) is associated with an 8.94-fold increase in the risk of RA (p = 0.044) and an 8.78-fold increase in BD (p = 0.040) but a 0.10 g/cm2 increase in bone density related to OP (p = 0.014). Each per-unit increase in copper is associated with a 0.87-fold increase in the risk of AD (p = 0.050) and BD (p = 0.010). In addition, there is suggestive evidence that calcium is positively correlated (OR = 1.36, p = 0.030) and iron is negatively correlated with T2D risk (OR = 0.89, p = 0.010); both magnesium (OR = 0.26, p = 0.013) and iron (OR = 0.71, p = 0.047) are negatively correlated with gout risk. In the sensitivity analysis, causal estimation is not affected by pleiotropy. This study supports the long-standing hypothesis that magnesium supplementation can increase RA and BD risks and decrease OP risk and that copper intake can reduce AD and BD risks. This study will be helpful to address some controversial debates on the relationships between minerals and chronic diseases.
Collapse
|
547
|
Suzuki K, Akiyama M, Ishigaki K, Kanai M, Hosoe J, Shojima N, Hozawa A, Kadota A, Kuriki K, Naito M, Tanno K, Ishigaki Y, Hirata M, Matsuda K, Iwata N, Ikeda M, Sawada N, Yamaji T, Iwasaki M, Ikegawa S, Maeda S, Murakami Y, Wakai K, Tsugane S, Sasaki M, Yamamoto M, Okada Y, Kubo M, Kamatani Y, Horikoshi M, Yamauchi T, Kadowaki T. Identification of 28 new susceptibility loci for type 2 diabetes in the Japanese population. Nat Genet 2019; 51:379-386. [PMID: 30718926 DOI: 10.1038/s41588-018-0332-4] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022]
Abstract
To understand the genetics of type 2 diabetes in people of Japanese ancestry, we conducted A meta-analysis of four genome-wide association studies (GWAS; 36,614 cases and 155,150 controls of Japanese ancestry). We identified 88 type 2 diabetes-associated loci (P < 5.0 × 10-8) with 115 independent signals (P < 5.0 × 10-6), of which 28 loci with 30 signals were novel. Twenty-eight missense variants were in linkage disequilibrium (r2 > 0.6) with the lead variants. Among the 28 missense variants, three previously unreported variants had distinct minor allele frequency (MAF) spectra between people of Japanese and European ancestry (MAFJPN > 0.05 versus MAFEUR < 0.01), including missense variants in genes related to pancreatic acinar cells (GP2) and insulin secretion (GLP1R). Transethnic comparisons of the molecular pathways identified from the GWAS results highlight both ethnically shared and heterogeneous effects of a series of pathways on type 2 diabetes (for example, monogenic diabetes and beta cells).
Collapse
Affiliation(s)
- Ken Suzuki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Centre for Integrative Medical Sciences, Yokohama, Japan.,Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jun Hosoe
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuhiro Shojima
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Hozawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.,Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Aya Kadota
- Center for Epidemiologic Research in Asia, Shiga University of Medical Science, Otsu, Japan.,Department of Public Health, Shiga University of Medical Science, Otsu, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Oral Epidemiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kozo Tanno
- Division of Clinical Research and Epidemiology, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Iwate, Japan.,Department of Hygiene and Preventive Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Yasushi Ishigaki
- Division of Innovation & Education, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Iwate, Japan.,Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Makoto Hirata
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Matsuda
- Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Aichi, Japan
| | - Masashi Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Aichi, Japan
| | - Norie Sawada
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Taiki Yamaji
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Motoki Iwasaki
- Division of Epidemiology, Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Shiro Maeda
- Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Centre for Integrative Medical Sciences, Yokohama, Japan.,Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan.,Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichiro Tsugane
- Center for Public Health Sciences, National Cancer Center, Tokyo, Japan
| | - Makoto Sasaki
- Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Iwate, Japan.,Division of Ultrahigh Field MRI, Institute for Biomedical Sciences, Iwate Medical University, Iwate, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan.,Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan.,Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan. .,Kyoto-McGill International Collaborative School in Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Momoko Horikoshi
- Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Centre for Integrative Medical Sciences, Yokohama, Japan.
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. .,Department of Prevention of Diabetes and Lifestyle-Related Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. .,Department of Metabolism and Nutrition, Mizonokuchi Hospital, Faculty of Medicine, Teikyo University, Tokyo, Japan.
| |
Collapse
|
548
|
Li Z, Chen P, Chen J, Xu Y, Wang Q, Li X, Li C, He L, Shi Y. Glucose and Insulin-Related Traits, Type 2 Diabetes and Risk of Schizophrenia: A Mendelian Randomization Study. EBioMedicine 2018; 34:182-188. [PMID: 30100396 PMCID: PMC6116472 DOI: 10.1016/j.ebiom.2018.07.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The link between schizophrenia and diabetes mellitus is well established by observational studies; however, the cause-effect relationship remains unclear. METHODS Here, we conducted Mendelian randomization analyses to assess a causal relationship of the genetic variants related to elevated fasting glucose levels, hemoglobin A1c (HbA1c), fasting insulin levels, and type 2 diabetes with the risk of schizophrenia. The analyses were performed using summary statistics obtained for the variants identified from the genome-wide association meta-analyses of fasting glucose levels (up to 133,010 individuals), HbA1c (up to 153,377 individuals), fasting insulin levels (up to 108,557 individuals), type 2 diabetes (up to 659,316 individuals), and schizophrenia (up to 108,341 individuals). The association between each variant and schizophrenia was weighted by its association with each studied condition, and estimates were combined using an inverse-variance weighted meta-analysis. FINDINGS Using information from thirteen variants related to fasting insulin levels, the causal effect of fasting insulin levels increases (per 1-SD) on the risk of schizophrenia was estimated at an odds ratio (OR) of 2·33 (p = 0·001), which is consistent with findings from the observational studies. The fasting glucose associated single nucleotide polymorphisms (SNPs) had no effect on the risk of schizophrenia in Europeans and East Asians (p > 0·05). Nonsignificant effects on the risk of schizophrenia was observed with raised HbA1c and type 2 diabetes, and consistent estimates were obtained across different populations. INTERPRETATION Our results suggest a causal role of elevated fasting insulin levels in schizophrenia pathogenesis.
Collapse
Affiliation(s)
- Zhiqiang Li
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, PR China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, PR China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China.
| | - Peng Chen
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, No. 45 Chaoyang Xi Road, Changchun 130021, PR China; College of Basic Medical Sciences, Jilin University, No. 126 Xinmin Street, Changchun 130021, PR China; National Institute of Digestive, Diabetes and Kidney Diseases, National Institutes of Health, 445 N 5th St, Phoenix, AZ 85004, USA
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, PR China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, No. 600 Wanping Nan Road, Shanghai 200030, PR China
| | - Yifeng Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, No. 600 Wanping Nan Road, Shanghai 200030, PR China
| | - Qingzhong Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Disease, The Metabolic Disease Institute of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, PR China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China
| | - Yongyong Shi
- The Affiliated Hospital of Qingdao University, The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, PR China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Shanghai 200240, PR China; Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, No. 1954 Huashan Road, Shanghai 200030, PR China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, No. 600 Wanping Nan Road, Shanghai 200030, PR China.
| |
Collapse
|