251
|
Li M, Li Q, Li CG, Guo M, Xu JM, Tang YY, Zhao QS, Hu YH, Cheng ZF, Zhang JC. Genetic polymorphisms in the PDZK1 gene and susceptibility to gout in male Han Chinese: a case-control study. Int J Clin Exp Med 2015; 8:13911-13918. [PMID: 26550347 PMCID: PMC4613032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/28/2015] [Indexed: 06/05/2023]
Abstract
PDZK1 acts as a scaffolding protein for a large variety of transporter and regulatory proteins, and has been identified in the kidney. The PDZK1 locus has been determined to be associated with the serum urate concentration. However, the evidence supporting this protein's association with gout is equivocal. In the current study, we investigated the association between two single nucleotide polymorphisms (SNPs) (rs12129861 and rs1967017) in the PDZK1 gene with gout in a male Chinese Han population. A total of 824 subjects were enrolled in this case-control study (400 gout cases and 424 controls). PDZK1 genotyping was carried out by polymerase chain reaction (PCR) and ligase detection reaction (LDR) assays methods. The relationships were evaluated using the pooled odds ratios (ORs) and their 95 % confidence intervals (CI). The results of our case-control study demonstrated that the gout and control groups exhibited significant differences in the distribution of genotypes at rs12129861 (OR = 0.727, P = 0.015) and rs1967017 (OR = 0.705, P = 0.016), suggesting that PDZK1 genetic polymorphisms were associated with increased risks of gout in male Han Chinese. However, there were no differences in the distribution of genotypes at rs12129861 (odds ratio (OR) = 0.744, P > 0.05) and rs1967017 (OR = 0.706, P > 0.05) in patients with gout with kidney stones and without kidney stones.
Collapse
Affiliation(s)
- Ming Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University37 Yiyuan Street, Harbin, Heilongjiang, China
| | - Qiang Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University37 Yiyuan Street, Harbin, Heilongjiang, China
| | - Chang-Gui Li
- Shandong Provincial Key Laboratory of Metabolic Disease, The Affiliated Hospital of Medical College, Qingdao UniversityQingdao, China
| | - Min Guo
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University148 Baojian Road, Harbin, Heilongjiang, China
| | - Jin-Mei Xu
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University148 Baojian Road, Harbin, Heilongjiang, China
| | - Ying-Ying Tang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University148 Baojian Road, Harbin, Heilongjiang, China
| | - Qing-Song Zhao
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University148 Baojian Road, Harbin, Heilongjiang, China
| | - Yu-Hua Hu
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University148 Baojian Road, Harbin, Heilongjiang, China
| | - Zhi-Feng Cheng
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University148 Baojian Road, Harbin, Heilongjiang, China
| | - Jin-Chao Zhang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University37 Yiyuan Street, Harbin, Heilongjiang, China
| |
Collapse
|
252
|
Genetic Variants Associated with Lipid Profiles in Chinese Patients with Type 2 Diabetes. PLoS One 2015; 10:e0135145. [PMID: 26252223 PMCID: PMC4529182 DOI: 10.1371/journal.pone.0135145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/17/2015] [Indexed: 11/30/2022] Open
Abstract
Dyslipidemia is a strong risk factor for cardiovascular disease among patients with type 2 diabetes (T2D). The aim of this study was to identify lipid-related genetic variants in T2D patients of Han Chinese ancestry. Among 4,908 Chinese T2D patients who were not taking lipid-lowering medications, single nucleotide polymorphisms (SNPs) in seven genes previously found to be associated with lipid traits in genome-wide association studies conducted in populations of European ancestry (ABCA1, GCKR, BAZ1B, TOMM40, DOCK7, HNF1A, and HNF4A) were genotyped. After adjusting for multiple covariates, SNPs in ABCA1, GCKR, BAZ1B, TOMM40, and HNF1A were identified as significantly associated with triglyceride levels in T2D patients (P < 0.05). The associations between the SNPs in ABCA1 (rs3890182), GCKR (rs780094), and BAZ1B (rs2240466) remained significant even after correction for multiple testing (P = 8.85×10−3, 7.88×10−7, and 2.03×10−6, respectively). BAZ1B (rs2240466) also was associated with the total cholesterol level (P = 4.75×10−2). In addition, SNP rs157580 in TOMM40 was associated with the low-density lipoprotein cholesterol level (P = 6.94×10−3). Our findings confirm that lipid-related genetic loci are associated with lipid profiles in Chinese patients with type 2 diabetes.
Collapse
|
253
|
Chandratre P, Mallen CD, Roddy E, Liddle J, Richardson J. "You want to get on with the rest of your life": a qualitative study of health-related quality of life in gout. Clin Rheumatol 2015; 35:1197-205. [PMID: 26245722 PMCID: PMC4844632 DOI: 10.1007/s10067-015-3039-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/14/2015] [Accepted: 07/23/2015] [Indexed: 11/30/2022]
Abstract
The objective of the study is to examine the impact of gout and its treatments on health-related quality of life (HRQOL) using focus group interviews. From the baseline phase of a cohort study of HRQOL in gout, 17 participants (15 males, mean age 71 years) with varying attack frequency and treatment with and without allopurinol participated in one of four focus group interviews. All interviews were audio-recorded and transcribed verbatim. Data was analysed thematically. Physical and psychosocial HRQOL in gout was affected by characteristics of acute gout (particularly the unpredictable nature of attacks, location of joint involved in an attack, pain and modifications in lifestyle), lack of understanding of gout by others (association with unhealthy lifestyle, symptoms ridiculed as non-severe and non-serious) as well as participants (not considered a disease) and the lack of information provided by physicians (about causes and pharmacological as well as non-pharmacological treatments of gout). Participants emphasised the impact of acute attacks of gout and prioritised dietary modifications and treatment of acute attacks over long-term urate-lowering therapy. Characteristics of acute gout, lack of understanding and information about gout and its treatments perpetuate poor HRQOL. HRQOL (maintenance of usual diet and reduced frequency of attacks) was associated with urate-lowering treatment. Better patient, public and practitioner education about gout being a chronic condition associated with co-morbidities and poor HRQOL may improve understanding and long-term treatment of gout.
Collapse
Affiliation(s)
- Priyanka Chandratre
- Research Institute of Primary Care and Health Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Christian D Mallen
- Research Institute of Primary Care and Health Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Edward Roddy
- Research Institute of Primary Care and Health Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Jennifer Liddle
- Research Institute of Primary Care and Health Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Jane Richardson
- Research Institute of Primary Care and Health Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| |
Collapse
|
254
|
Sluijs I, Holmes MV, van der Schouw YT, Beulens JWJ, Asselbergs FW, Huerta JM, Palmer TM, Arriola L, Balkau B, Barricarte A, Boeing H, Clavel-Chapelon F, Fagherazzi G, Franks PW, Gavrila D, Kaaks R, Khaw KT, Kühn T, Molina-Montes E, Mortensen LM, Nilsson PM, Overvad K, Palli D, Panico S, Quirós JR, Rolandsson O, Sacerdote C, Sala N, Schmidt JA, Scott RA, Sieri S, Slimani N, Spijkerman AMW, Tjonneland A, Travis RC, Tumino R, van der A DL, Sharp SJ, Forouhi NG, Langenberg C, Riboli E, Wareham NJ. A Mendelian Randomization Study of Circulating Uric Acid and Type 2 Diabetes. Diabetes 2015; 64:3028-36. [PMID: 25918230 PMCID: PMC6284788 DOI: 10.2337/db14-0742] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 04/07/2015] [Indexed: 01/08/2023]
Abstract
We aimed to investigate the causal effect of circulating uric acid concentrations on type 2 diabetes risk. A Mendelian randomization study was performed using a genetic score with 24 uric acid-associated loci. We used data of the European Prospective Investigation into Cancer and Nutrition (EPIC)-InterAct case-cohort study, comprising 24,265 individuals of European ancestry from eight European countries. During a mean (SD) follow-up of 10 (4) years, 10,576 verified incident case subjects with type 2 diabetes were ascertained. Higher uric acid was associated with a higher diabetes risk after adjustment for confounders, with a hazard ratio (HR) of 1.20 (95% CI 1.11, 1.30) per 59.48 µmol/L (1 mg/dL) uric acid. The genetic score raised uric acid by 17 µmol/L (95% CI 15, 18) per SD increase and explained 4% of uric acid variation. By using the genetic score to estimate the unconfounded effect, we found that a 59.48 µmol/L higher uric acid concentration did not have a causal effect on diabetes (HR 1.01 [95% CI 0.87, 1.16]). Including data from the Diabetes Genetics Replication And Meta-analysis (DIAGRAM) consortium, increasing our dataset to 41,508 case subjects with diabetes, the summary odds ratio estimate was 0.99 (95% CI 0.92, 1.06). In conclusion, our study does not support a causal effect of circulating uric acid on diabetes risk. Uric acid-lowering therapies may therefore not be beneficial in reducing diabetes risk.
Collapse
Affiliation(s)
- Ivonne Sluijs
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michael V Holmes
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, U.K. Division of Transplantation and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joline W J Beulens
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Folkert W Asselbergs
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands Department of Cardiology, Heart Long Institute, University Medical Center Utrecht, Utrecht, the Netherlands Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| | - José María Huerta
- Department of Epidemiology, Murcia Regional Health Council, Murcia, Spain CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Tom M Palmer
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, U.K
| | - Larraitz Arriola
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain Public Health Division of Gipuzkoa, San Sebastian, Spain Instituto BIO-Donostia, Basque Government, San Sebastian, Spain
| | - Beverley Balkau
- Inserm, Center for Research in Epidemiology and Population Health (CESP), U1018, Villejuif, France Université Paris-Sud, UMRS 1018, Villejuif, France
| | - Aurelio Barricarte
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain Navarre Public Health Institute (ISPN), Pamplona, Spain
| | - Heiner Boeing
- German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Françoise Clavel-Chapelon
- Inserm, Center for Research in Epidemiology and Population Health (CESP), U1018, Villejuif, France Université Paris-Sud, UMRS 1018, Villejuif, France
| | - Guy Fagherazzi
- Inserm, Center for Research in Epidemiology and Population Health (CESP), U1018, Villejuif, France Université Paris-Sud, UMRS 1018, Villejuif, France
| | - Paul W Franks
- Lund University, Malmö, Sweden Umeå University, Umeå, Sweden
| | - Diana Gavrila
- Department of Epidemiology, Murcia Regional Health Council, Murcia, Spain CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Rudolf Kaaks
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Tilman Kühn
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Esther Molina-Montes
- CIBER Epidemiología y Salud Pública (CIBERESP), Spain Andalusian School of Public Health, Granada, Spain
| | - Lotte Maxild Mortensen
- Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | | | - Kim Overvad
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark Aalborg University Hospital, Aalborg, Denmark
| | - Domenico Palli
- Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | | | | | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Citta' della Salute e della Scienza Hospital, University of Turin and Center for Cancer Prevention (CPO), Turin, Italy Human Genetics Foundation (HuGeF), Turin, Italy
| | - Núria Sala
- Unit of Nutrition, Environment and Cancer, Cancer Epidemiology Research Program, and Translational Research Laboratory, Catalan Institute of Oncology (IDIBELL), Barcelona, Spain
| | - Julie A Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | | | - Nadia Slimani
- International Agency for Research on Cancer, Lyon, France
| | | | | | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | | | - Daphne L van der A
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Stephen J Sharp
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge, Cambridge, U.K
| | | | - Elio Riboli
- School of Public Health, Imperial College London, U.K
| | | |
Collapse
|
255
|
Uric Acid Produces an Inflammatory Response through Activation of NF-κB in the Hypothalamus: Implications for the Pathogenesis of Metabolic Disorders. Sci Rep 2015; 5:12144. [PMID: 26179594 PMCID: PMC4503982 DOI: 10.1038/srep12144] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/18/2015] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies have shown that an elevated uric acid (UA) level predicts the development of metabolic syndrome and diabetes; however, there is no direct evidence of this, and the underlying mechanism remains unclear. Here, we showed that a high-UA diet triggered the expression of pro-inflammatory cytokines, activated the NF-κB pathway, and increased gliosis in the hypothalamus. Intracerebroventricular injection of UA induced hypothalamic inflammation and reactive gliosis, whereas these effects were markedly ameliorated by the inhibition of NF-κB. Moreover, magnetic resonance imaging confirmed that hyperuricemia in rodents and humans was associated with gliosis in the mediobasal hypothalamus. Importantly, the rats administered UA exhibited dyslipidemia and glucose intolerance, which were probably mediated by hypothalamic inflammation and hypothalamic neuroendocrine alterations. These results suggest that UA can cause hypothalamic inflammation via NF-κB signaling. Our findings provide a potential therapeutic strategy for UA-induced metabolic disorders.
Collapse
|
256
|
Kastenmüller G, Raffler J, Gieger C, Suhre K. Genetics of human metabolism: an update. Hum Mol Genet 2015; 24:R93-R101. [PMID: 26160913 PMCID: PMC4572003 DOI: 10.1093/hmg/ddv263] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/06/2015] [Indexed: 01/01/2023] Open
Abstract
Genome-wide association studies with metabolomics (mGWAS) identify genetically influenced metabotypes (GIMs), their ensemble defining the heritable part of every human's metabolic individuality. Knowledge of genetic variation in metabolism has many applications of biomedical and pharmaceutical interests, including the functional understanding of genetic associations with clinical end points, design of strategies to correct dysregulations in metabolic disorders and the identification of genetic effect modifiers of metabolic disease biomarkers. Furthermore, it has been shown that GIMs provide testable hypotheses for functional genomics and metabolomics and for the identification of novel gene functions and metabolite identities. mGWAS with growing sample sizes and increasingly complex metabolic trait panels are being conducted, allowing for more comprehensive and systems-based downstream analyses. The generated large datasets of genetic associations can now be mined by the biomedical research community and provide valuable resources for hypothesis-driven studies. In this review, we provide a brief summary of the key aspects of mGWAS, followed by an update of recently published mGWAS. We then discuss new approaches of integrating and exploring mGWAS results and finish by presenting selected applications of GIMs in recent studies.
Collapse
Affiliation(s)
- Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, German Center for Diabetes Research, Neuherberg, Germany and
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany and Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, Department of Physiology and Biophysics, Weill Cornell Medical College-Qatar, Doha, Qatar
| |
Collapse
|
257
|
Tsepilov YA, Shin SY, Soranzo N, Spector TD, Prehn C, Adamski J, Kastenmüller G, Wang-Sattler R, Strauch K, Gieger C, Aulchenko YS, Ried JS. Nonadditive Effects of Genes in Human Metabolomics. Genetics 2015; 200:707-18. [PMID: 25977471 PMCID: PMC4512538 DOI: 10.1534/genetics.115.175760] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 05/04/2015] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies (GWAS) are widely applied to analyze the genetic effects on phenotypes. With the availability of high-throughput technologies for metabolite measurements, GWAS successfully identified loci that affect metabolite concentrations and underlying pathways. In most GWAS, the effect of each SNP on the phenotype is assumed to be additive. Other genetic models such as recessive, dominant, or overdominant were considered only by very few studies. In contrast to this, there are theories that emphasize the relevance of nonadditive effects as a consequence of physiologic mechanisms. This might be especially important for metabolites because these intermediate phenotypes are closer to the underlying pathways than other traits or diseases. In this study we analyzed systematically nonadditive effects on a large panel of serum metabolites and all possible ratios (22,801 total) in a population-based study [Cooperative Health Research in the Region of Augsburg (KORA) F4, N = 1,785]. We applied four different 1-degree-of-freedom (1-df) tests corresponding to an additive, dominant, recessive, and overdominant trait model as well as a genotypic model with two degree-of-freedom (2-df) that allows a more general consideration of genetic effects. Twenty-three loci were found to be genome-wide significantly associated (Bonferroni corrected P ≤ 2.19 × 10(-12)) with at least one metabolite or ratio. For five of them, we show the evidence of nonadditive effects. We replicated 17 loci, including 3 loci with nonadditive effects, in an independent study (TwinsUK, N = 846). In conclusion, we found that most genetic effects on metabolite concentrations and ratios were indeed additive, which verifies the practice of using the additive model for analyzing SNP effects on metabolites.
Collapse
Affiliation(s)
- Yakov A Tsepilov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia Novosibirsk State University, 630090 Novosibirsk, Russia Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - So-Youn Shin
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, United Kingdom MRC Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, BS8 1TH, United Kingdom
| | - Nicole Soranzo
- Human Genetics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, CB10 1HH, United Kingdom Department of Haematology, University of Cambridge, Cambridge, CB2 0AH, United Kingdom
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, WC2R 2LS, United Kingdom
| | - Cornelia Prehn
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany Institute of Experimental Genetics, Life and Food Science Center Weihenstephan, Technische Universität München, 85354 Freising-Weihenstephan, Germany German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Gabi Kastenmüller
- Department of Twin Research and Genetic Epidemiology, King's College London, London, WC2R 2LS, United Kingdom Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Rui Wang-Sattler
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, 85764 Neuherberg, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Yurii S Aulchenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Janina S Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| |
Collapse
|
258
|
Tomi M, Eguchi H, Ozaki M, Tawara T, Nishimura S, Higuchi K, Maruyama T, Nishimura T, Nakashima E. Role of OAT4 in Uptake of Estriol Precursor 16α-Hydroxydehydroepiandrosterone Sulfate Into Human Placental Syncytiotrophoblasts From Fetus. Endocrinology 2015; 156:2704-12. [PMID: 25919187 DOI: 10.1210/en.2015-1130] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Estriol biosynthesis in human placenta requires the uptake of a fetal liver-derived estriol precursor, 16α-hydroxydehydroepiandrosterone sulfate (16α-OH DHEAS), by placental syncytiotrophoblasts at their basal plasma membrane (BM), which faces the fetal circulation. The aim of this work is to identify the transporter(s) mediating 16α-OH DHEAS uptake at the fetal side of syncytiotrophoblasts by using human placental BM-enriched vesicles and to examine the contribution of the putative transporter to estriol synthesis at the cellular level, using choriocarcinoma JEG-3 cells. Organic anion transporter (OAT)-4 and organic anion transporting polypeptide 2B1 proteins were enriched in human placental BM vesicles compared with crude membrane fraction. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was partially inhibited in the absence of sodium but was significantly increased in the absence of chloride and after preloading glutarate. Uptake of [(3)H]16α-OH DHEAS by BM vesicles was significantly inhibited by OAT4 substrates such as dehydroepiandrosterone sulfate, estrone-3-sulfate, and bromosulfophthalein but not by cyclosporin A, tetraethylammonium, p-aminohippuric acid, or cimetidine. These characteristics of vesicular [(3)H]16α-OH DHEAS uptake are in good agreement with those of human OAT4-transfected COS-7 cells as well as forskolin-differentiated JEG-3 cells. Estriol secretion from differentiated JEG-3 cells was detected when the cells were incubated with 16α-OH DHEAS for 8 hours but was inhibited in the presence of 50 μM bromosulfophthalein. Our results indicate that OAT4 at the BM of human placental syncytiotrophoblasts plays a predominant role in the uptake of 16α-OH DHEAS for placental estriol synthesis.
Collapse
Affiliation(s)
- Masatoshi Tomi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Hiromi Eguchi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Mayuko Ozaki
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tomohiro Tawara
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Sachika Nishimura
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Kei Higuchi
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tetsuo Maruyama
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Tomohiro Nishimura
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| | - Emi Nakashima
- Faculty of Pharmacy (M.T., H.E., M.O., T.T., S.N., K.H., T.N., E.N.), Keio University, Minato-ku 105-8512, Tokyo, Japan; School of Pharmaceutical Sciences (K.H.), Teikyo University, Itabashi-ku 173-8605, Tokyo, Japan; and Department of Obstetrics and Gynecology (T.M.), School of Medicine, Keio University, Shinjuku-ku 160-8512, Tokyo, Japan
| |
Collapse
|
259
|
Ogura J, Kuwayama K, Sasaki S, Kaneko C, Koizumi T, Yabe K, Tsujimoto T, Takeno R, Takaya A, Kobayashi M, Yamaguchi H, Iseki K. Reactive oxygen species derived from xanthine oxidase interrupt dimerization of breast cancer resistance protein, resulting in suppression of uric acid excretion to the intestinal lumen. Biochem Pharmacol 2015; 97:89-98. [PMID: 26119820 DOI: 10.1016/j.bcp.2015.06.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/22/2015] [Indexed: 02/04/2023]
Abstract
The prevalence of hyperuricemia/gout increases with aging. However, the effect of aging on function for excretion of uric acid to out of the body has not been clarified. We found that ileal uric acid clearance in middle-aged rats (11-12 months) was decreased compared with that in young rats (2 months). In middle-aged rats, xanthine oxidase (XO) activity in the ileum was significantly higher than that in young rats. Inosine-induced reactive oxygen species (ROS), which are derived from XO, also decreased ileal uric acid clearance. ROS derived from XO decreased the active homodimer level of breast cancer resistance protein (BCRP), which is a uric acid efflux transporter, in the ileum. Pre-administration of allopurinol recovered the BCRP homodimer level, resulting in the recovering ileal uric acid clearance. Moreover, we investigated the effects of ROS derived from XO on BCRP homodimer level directly in Caco-2 cells using hypoxanthine. Treatment with hypoxanthine decreased BCRP homodimer level. Treatment with hypoxanthine induced mitochondrial dysfunction, suggesting that the decreasing BCRP homodimer level might be caused by mitochondrial dysfunction. In conclusion, ROS derived from XO decrease BCRP homodimer level, resulting in suppression of function for uric acid excretion to the ileal lumen. ROS derived from XO may cause the suppression of function of the ileum for the excretion of uric acid with aging. The results of our study provide a new insight into the causes of increasing hyperuricemia/gout prevalence with aging.
Collapse
Affiliation(s)
- Jiro Ogura
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Kaori Kuwayama
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Shunichi Sasaki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Chihiro Kaneko
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Takahiro Koizumi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Keisuke Yabe
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Takashi Tsujimoto
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Reiko Takeno
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Atsushi Takaya
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Hiroaki Yamaguchi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Ken Iseki
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12-jo, Nishi-6-chome, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
260
|
Dalbeth N, Topless R, Flynn T, Cadzow M, Bolland MJ, Merriman TR. Mendelian randomization analysis to examine for a causal effect of urate on bone mineral density. J Bone Miner Res 2015; 30:985-91. [PMID: 25502344 DOI: 10.1002/jbmr.2434] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 11/05/2022]
Abstract
In observational studies, serum urate concentrations are positively associated with bone mineral density (BMD) and reduced risk of fragility fractures, raising the possibility that urate is a direct mediator of bone density. We used Mendelian randomization analysis to examine whether urate has a causal effect on BMD. We analyzed data from the Generation 3 cohort in the Framingham Heart Study (FHS) (N = 2501 total; 1265 male, 1236 female). A weighted genetic urate score was calculated using the SLC2A9, ABCG2, SLC17A1, SLC22A11, and SLC22A12 single-nucleotide polymorphisms (SNPs) that explains 3.4% of the variance in serum urate. Mendelian randomization analysis was performed using the two-stage least squares method with >80% power at α = 0.05 to detect an effect size equivalent to that observed in the ordinary least squares analysis between serum urate and total femur BMD. A strong association between serum urate and BMD was observed in the crude ordinary least squares analysis (total femur crude beta = 0.47, p = 1.7E-51). In the two-stage least squares analysis using the weighted genetic urate score as the instrumental variable, no significant relationship was observed between serum urate and BMD (total femur crude beta =-0.36, p = 0.06). Similar findings were observed in both the male and female subgroups, and there was no evidence for causality when individual SNPs were analyzed. Serum urate is strongly associated with BMD. However, controlling for confounders by Mendelian randomization analysis does not provide evidence that increased urate has a causal effect on increasing BMD.
Collapse
Affiliation(s)
- Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Ruth Topless
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Tanya Flynn
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Murray Cadzow
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Mark J Bolland
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
261
|
Zykova SN, Storhaug HM, Toft I, Chadban SJ, Jenssen TG, White SL. Cross-sectional analysis of nutrition and serum uric acid in two Caucasian cohorts: the AusDiab Study and the Tromsø study. Nutr J 2015; 14:49. [PMID: 25971955 PMCID: PMC4459487 DOI: 10.1186/s12937-015-0032-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 04/27/2015] [Indexed: 02/07/2023] Open
Abstract
Background Hyperuricemia can lead to gout, and may be a risk factor for cardiovascular events, hypertension, diabetes and renal disease. There is well-known link between gout and habitual intake of meat and seafood, however the association between hyperuricemia and micro-and macro-nutrient intake has not been established. Methods We studied associations between intakes of food categories, macro-and micronutrients and serum uric acid (SUA) levels in two cross-sectional surveys of Caucasian adults deriving from different food traditions: Australian Diabetes, Obesity and Lifestyle Study 1999/00 (n=9734, age 25–91) and Tromsø Study 4 1994/95 (n = 3031, age 25–69). Dietary intake was calculated from self-administered Food Frequency Questionnaires. In some analyses we stratified according to abdominal obesity status and gender. Results In both cohorts, lower levels of SUA were found in subjects with higher consumption of carbohydrates, calcium and vitamin B2, while higher fat intake was associated with higher SUA, after adjustment for age, body mass index, estimated glomerular filtration rate, physical activity, total energy intake, use of diuretics, presence of hypertension, diabetes and gout. Among individual food items, high consumption of dairy products, high-fibre bread, cereals and fruits were associated with lower SUA in most subject groups while consumption of meat, eggs, beer and spirits, but not wine, with elevated levels. Conclusions Healthy food choices with high intake of carbohydrates, dairy products, fiber and micronutrient-rich foods, and limited intake of fat, beer and spirits, might be recommended to prevent high SUA. Dietary factors seem to have qualitatively similar impact on SUA in obese and non-obese men and women from Australia and Norway. Electronic supplementary material The online version of this article (doi:10.1186/s12937-015-0032-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Svetlana N Zykova
- Clinical Research Department, University Hospital of North Norway, 9038, Tromsø, Norway.
| | - Hilde M Storhaug
- University of Tromsø-The Arctic University of Norway, Tromsø, Norway.
| | - Ingrid Toft
- Clinical Research Department, University Hospital of North Norway, 9038, Tromsø, Norway. .,University of Tromsø-The Arctic University of Norway, Tromsø, Norway.
| | - Steven J Chadban
- Royal Prince Alfred Hospital, Sydney, Australia. .,Sydney Medical School, University of Sydney, Sydney, Australia.
| | - Trond G Jenssen
- University of Tromsø-The Arctic University of Norway, Tromsø, Norway. .,Oslo University Hospital, Oslo, Norway.
| | - Sarah L White
- Sydney Medical School, University of Sydney, Sydney, Australia.
| |
Collapse
|
262
|
Genome-wide association analysis identifies three new risk loci for gout arthritis in Han Chinese. Nat Commun 2015; 6:7041. [PMID: 25967671 PMCID: PMC4479022 DOI: 10.1038/ncomms8041] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 03/26/2015] [Indexed: 12/31/2022] Open
Abstract
Gout is one of the most common types of inflammatory arthritis, caused by the deposition of monosodium urate crystals in and around the joints. Previous genome-wide association studies (GWASs) have identified many genetic loci associated with raised serum urate concentrations. However, hyperuricemia alone is not sufficient for the development of gout arthritis. Here we conduct a multistage GWAS in Han Chinese using 4,275 male gout patients and 6,272 normal male controls (1,255 cases and 1,848 controls were genome-wide genotyped), with an additional 1,644 hyperuricemic controls. We discover three new risk loci, 17q23.2 (rs11653176, P=1.36 × 10−13, BCAS3), 9p24.2 (rs12236871, P=1.48 × 10−10, RFX3) and 11p15.5 (rs179785, P=1.28 × 10−8, KCNQ1), which contain inflammatory candidate genes. Our results suggest that these loci are most likely related to the progression from hyperuricemia to inflammatory gout, which will provide new insights into the pathogenesis of gout arthritis. Raised serum urate levels are a risk factor for gout, a common form of inflammatory arthritis. Here Li et al. conduct a multistage genome-wide association study in a Han Chinese population and identify three novel loci likely associated with the progression from hyperuricemia to gout.
Collapse
|
263
|
Roberts RL, Stamp LK. Pharmacogenetic considerations in the treatment of gout. Pharmacogenomics 2015; 16:619-29. [PMID: 25876828 DOI: 10.2217/pgs.15.16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gout is one of the most common forms of arthritis and the prevalence is increasing. Management comprises rapid and effective control of the inflammation in acute gout and sustained urate lowering in the long term. Improving the outcomes for cheaper old drugs and for the increasing number of new, more expensive agents is an important clinical goal. The role of pharmacogenetics in predicting response and adverse events to gout therapies is of considerable interest. Currently, prospective screening is employed to detect HLA-B*5801 carriage and glucose-6-phosphate dehydrogenase deficiency, to minimize occurrence of allopurinol hypersensitivity and pegloticase-related hemolytic anemia. In the future it is likely that other genetic markers of drug response will make the transition to clinical practice to further improve the efficacy and safety of gout therapies. In this review, we will examine the potential clinical relevance of specific genetic variants in the management of gout.
Collapse
Affiliation(s)
- Rebecca L Roberts
- Department of Surgical Sciences, Dunedin School of Medicine, Dunedin, New Zealand
| | | |
Collapse
|
264
|
Abstract
Genome-wide association studies that scan the genome for common genetic variants associated with phenotype have greatly advanced medical knowledge. Hyperuricemia is no exception, with 28 loci identified. However, genetic control of pathways determining gout in the presence of hyperuricemia is still poorly understood. Two important pathways determining hyperuricemia have been confirmed (renal and gut excretion of uric acid with glycolysis now firmly implicated). Major urate loci are SLC2A9 and ABCG2. Recent studies show that SLC2A9 is involved in renal and gut excretion of uric acid and is implicated in antioxidant defense. Although etiological variants at SLC2A9 are yet to be identified, it is clear that considerable genetic complexity exists at the SLC2A9 locus, with multiple statistically independent genetic variants and local epistatic interactions. The positions of implicated genetic variants within or near chromatin regions involved in transcriptional control suggest that this mechanism (rather than structural changes in SLC2A9) is important in regulating the activity of SLC2A9. ABCG2 is involved primarily in extra-renal uric acid under-excretion with the etiological variant influencing expression. At the other 26 loci, probable causal genes can be identified at three (PDZK1, SLC22A11, and INHBB) with strong candidates at a further 10 loci. Confirmation of the causal gene will require a combination of re-sequencing, trans-ancestral mapping, and correlation of genetic association data with expression data. As expected, the urate loci associate with gout, although inconsistent effect sizes for gout require investigation. Finally, there has been no genome-wide association study using clinically ascertained cases to investigate the causes of gout in the presence of hyperuricemia. In such a study, use of asymptomatic hyperurcemic controls would be expected to increase the ability to detect genetic associations with gout.
Collapse
Affiliation(s)
- Tony R Merriman
- Department of Biochemistry, University of Otago, Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
265
|
A polymorphism in the major gene regulating serum uric acid associates with clinic SBP and the white-coat effect in a family-based study. J Hypertens 2015; 32:1621-8; discussion 1628. [PMID: 24805955 DOI: 10.1097/hjh.0000000000000224] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Hyperuricemia associates with hypertension, but it is uncertain whether this relationship is causal in nature. Glucose transporter 9 (GLUT9) gene is a major genetic determinant of plasma uric acid levels in humans. Since polymorphisms are randomly distributed at mating (Mendelian randomization), studies based on GLUT9 polymorphisms may provide unconfounded assessment of the nature of the link between uric acid and hypertension. METHODS We tested the association between uric acid, the rs734553 polymorphism of the GLUT9 gene and arterial pressure in a family-based study including 449 individuals in a genetically homogenous population in Southern Italy. RESULTS Serum uric acid levels were strongly associated (P < 0.001) with all components of clinic and 24-h ambulatory blood pressures (BPs). However, only clinic SBP and the white-coat effect (the difference in clinic systolic and daytime systolic ambulatory blood pressure monitoring) associations remained significant after adjustment for classical risk factor and the estimated glomerular filtration rate. Serum uric acid was strongly associated with the risk allele (T) of the rs734553 polymorphism (P < 0.001). Furthermore, TT individuals showed higher clinic SBP (129 + SEM 1 mmHg) than GT (125 + 1 mmHg) and GG individuals (122 + 3 mmHg), as well as a higher white-coat effect (P = 0.02), confirming that the association between uric acid and these BP components is unconfounded by environmental risk factors. CONCLUSION Results in this family-based study are compatible with the hypothesis that uric acid is a causal risk factor for hypertension. Trials testing uric acid-lowering interventions are needed to definitively establish the causal implication of hyperuricemia in human hypertension. [Corrected]
Collapse
|
266
|
Raimondo A, Rees MG, Gloyn AL. Glucokinase regulatory protein: complexity at the crossroads of triglyceride and glucose metabolism. Curr Opin Lipidol 2015; 26:88-95. [PMID: 25692341 PMCID: PMC4422901 DOI: 10.1097/mol.0000000000000155] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE OF REVIEW Glucokinase regulator (GCKR) encodes glucokinase regulatory protein (GKRP), a hepatocyte-specific inhibitor of the glucose-metabolizing enzyme glucokinase (GCK). Genome-wide association studies have identified a common coding variant within GCKR associated with multiple metabolic traits. This review focuses on recent insights into the critical role of GKRP in hepatic glucose metabolism that have stemmed from the study of human genetics. This knowledge has improved our understanding of glucose and lipid physiology and informed the development of targeted molecular therapeutics for diabetes. RECENT FINDINGS Rare GCKR variants have effects on GKRP expression, localization, and activity. These variants are collectively associated with hypertriglyceridaemia but are not causal. Crystal structures of GKRP and the GCK-GKRP complex have been solved, providing greater insight into the molecular interactions between these proteins. Finally, small molecules have been identified that directly bind GKRP and reduce blood glucose levels in rodent models of diabetes. SUMMARY GCKR variants across the allelic spectrum have effects on glucose and lipid homeostasis. Functional analysis has highlighted numerous molecular mechanisms for GKRP dysfunction. Hepatocyte-specific GCK activation via small molecule GKRP inhibition may be a new avenue for type 2 diabetes treatment, particularly considering evidence indicating GKRP loss-of-function alone does not cause hypertriglyceridaemia.
Collapse
Affiliation(s)
- Anne Raimondo
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Matthew G. Rees
- Center for the Science of Therapeutics, Broad Institute, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Broad Institute, Cambridge, Massachusetts, USA
| | - Anna L. Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, ORH Trust, OCDEM, Churchill Hospital, Oxford, UK
| |
Collapse
|
267
|
|
268
|
Huffman JE, Albrecht E, Teumer A, Mangino M, Kapur K, Johnson T, Kutalik Z, Pirastu N, Pistis G, Lopez LM, Haller T, Salo P, Goel A, Li M, Tanaka T, Dehghan A, Ruggiero D, Malerba G, Smith AV, Nolte IM, Portas L, Phipps-Green A, Boteva L, Navarro P, Johansson A, Hicks AA, Polasek O, Esko T, Peden JF, Harris SE, Murgia F, Wild SH, Tenesa A, Tin A, Mihailov E, Grotevendt A, Gislason GK, Coresh J, D'Adamo P, Ulivi S, Vollenweider P, Waeber G, Campbell S, Kolcic I, Fisher K, Viigimaa M, Metter JE, Masciullo C, Trabetti E, Bombieri C, Sorice R, Döring A, Reischl E, Strauch K, Hofman A, Uitterlinden AG, Waldenberger M, Wichmann HE, Davies G, Gow AJ, Dalbeth N, Stamp L, Smit JH, Kirin M, Nagaraja R, Nauck M, Schurmann C, Budde K, Farrington SM, Theodoratou E, Jula A, Salomaa V, Sala C, Hengstenberg C, Burnier M, Mägi R, Klopp N, Kloiber S, Schipf S, Ripatti S, Cabras S, Soranzo N, Homuth G, Nutile T, Munroe PB, Hastie N, Campbell H, Rudan I, Cabrera C, Haley C, Franco OH, Merriman TR, Gudnason V, Pirastu M, Penninx BW, Snieder H, Metspalu A, Ciullo M, Pramstaller PP, van Duijn CM, et alHuffman JE, Albrecht E, Teumer A, Mangino M, Kapur K, Johnson T, Kutalik Z, Pirastu N, Pistis G, Lopez LM, Haller T, Salo P, Goel A, Li M, Tanaka T, Dehghan A, Ruggiero D, Malerba G, Smith AV, Nolte IM, Portas L, Phipps-Green A, Boteva L, Navarro P, Johansson A, Hicks AA, Polasek O, Esko T, Peden JF, Harris SE, Murgia F, Wild SH, Tenesa A, Tin A, Mihailov E, Grotevendt A, Gislason GK, Coresh J, D'Adamo P, Ulivi S, Vollenweider P, Waeber G, Campbell S, Kolcic I, Fisher K, Viigimaa M, Metter JE, Masciullo C, Trabetti E, Bombieri C, Sorice R, Döring A, Reischl E, Strauch K, Hofman A, Uitterlinden AG, Waldenberger M, Wichmann HE, Davies G, Gow AJ, Dalbeth N, Stamp L, Smit JH, Kirin M, Nagaraja R, Nauck M, Schurmann C, Budde K, Farrington SM, Theodoratou E, Jula A, Salomaa V, Sala C, Hengstenberg C, Burnier M, Mägi R, Klopp N, Kloiber S, Schipf S, Ripatti S, Cabras S, Soranzo N, Homuth G, Nutile T, Munroe PB, Hastie N, Campbell H, Rudan I, Cabrera C, Haley C, Franco OH, Merriman TR, Gudnason V, Pirastu M, Penninx BW, Snieder H, Metspalu A, Ciullo M, Pramstaller PP, van Duijn CM, Ferrucci L, Gambaro G, Deary IJ, Dunlop MG, Wilson JF, Gasparini P, Gyllensten U, Spector TD, Wright AF, Hayward C, Watkins H, Perola M, Bochud M, Kao WHL, Caulfield M, Toniolo D, Völzke H, Gieger C, Köttgen A, Vitart V. Modulation of genetic associations with serum urate levels by body-mass-index in humans. PLoS One 2015; 10:e0119752. [PMID: 25811787 PMCID: PMC4374966 DOI: 10.1371/journal.pone.0119752] [Show More Authors] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 02/03/2015] [Indexed: 11/17/2022] Open
Abstract
We tested for interactions between body mass index (BMI) and common genetic variants affecting serum urate levels, genome-wide, in up to 42569 participants. Both stratified genome-wide association (GWAS) analyses, in lean, overweight and obese individuals, and regression-type analyses in a non BMI-stratified overall sample were performed. The former did not uncover any novel locus with a major main effect, but supported modulation of effects for some known and potentially new urate loci. The latter highlighted a SNP at RBFOX3 reaching genome-wide significant level (effect size 0.014, 95% CI 0.008-0.02, Pinter= 2.6 x 10-8). Two top loci in interaction term analyses, RBFOX3 and ERO1LB-EDARADD, also displayed suggestive differences in main effect size between the lean and obese strata. All top ranking loci for urate effect differences between BMI categories were novel and most had small magnitude but opposite direction effects between strata. They include the locus RBMS1-TANK (men, Pdifflean-overweight= 4.7 x 10-8), a region that has been associated with several obesity related traits, and TSPYL5 (men, Pdifflean-overweight= 9.1 x 10-8), regulating adipocytes-produced estradiol. The top-ranking known urate loci was ABCG2, the strongest known gout risk locus, with an effect halved in obese compared to lean men (Pdifflean-obese= 2 x 10-4). Finally, pathway analysis suggested a role for N-glycan biosynthesis as a prominent urate-associated pathway in the lean stratum. These results illustrate a potentially powerful way to monitor changes occurring in obesogenic environment.
Collapse
Affiliation(s)
- Jennifer E Huffman
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexander Teumer
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Massimo Mangino
- King's College London, St. Thomas' Hospital Campus, London, United Kingdom
| | - Karen Kapur
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Toby Johnson
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Zoltán Kutalik
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nicola Pirastu
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy; University of Trieste, Trieste, Italy
| | - Giorgio Pistis
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Lorna M Lopez
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Perttu Salo
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Anuj Goel
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Toshiko Tanaka
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Abbas Dehghan
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Giovanni Malerba
- Biology and Genetics section, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Albert V Smith
- Icelandic Heart Association Research Institute, Kopavogur, Iceland; University of Iceland, Reykjavik, Iceland
| | - Ilja M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Laura Portas
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | | | - Lora Boteva
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Pau Navarro
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Asa Johansson
- Uppsala Clinical Research Center, Uppsala University Hospital, Upsalla, Sweden; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, 751 85, Sweden
| | - Andrew A Hicks
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy; Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Croatia, Soltanska 2, Split, 21000, Croatia
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia; Broad Institute, Cambridge, MA, United States of America; Children's Hospital Boston, Boston, MA, United States of America
| | - John F Peden
- Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom; Medical Genetics Section, University of Edinburgh Centre for Genomics and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | - Federico Murgia
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Sarah H Wild
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Albert Tenesa
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom; Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | | | - Anne Grotevendt
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Gauti K Gislason
- Icelandic Heart Association Research Institute, Kopavogur, Iceland
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America; Welch Center for Prevention, Epidemiology and Clinical Research, John Hopkins University, Baltimore, MD, United States of America
| | - Pio D'Adamo
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy; University of Trieste, Trieste, Italy
| | - Sheila Ulivi
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Gerard Waeber
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Susan Campbell
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Ivana Kolcic
- Faculty of Medicine, University of Split, Croatia, Soltanska 2, Split, 21000, Croatia
| | - Krista Fisher
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Margus Viigimaa
- Tallinn University of Technology, Department of Biomedical Engineering, Chair of Medical Physics, Tallinn, Estonia; Centre of Cardiology, North Estonia Medical Centre, Tallinn, Estonia
| | - Jeffrey E Metter
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Corrado Masciullo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Elisabetta Trabetti
- Biology and Genetics section, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Cristina Bombieri
- Biology and Genetics section, Department of Life and Reproduction Sciences, University of Verona, Verona, Italy
| | - Rossella Sorice
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Angela Döring
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Eva Reischl
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | - Albert Hofman
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andre G Uitterlinden
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Melanie Waldenberger
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - H-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-University, Munich, Germany; Klinikum Grosshadern, Munich, Germany
| | - Gail Davies
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alan J Gow
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola Dalbeth
- Bone and Joint Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Lisa Stamp
- Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Johannes H Smit
- Department of Psychiatry/EMGO Institute, VU University Medical Centre, Amsterdam, the Netherlands
| | - Mirna Kirin
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Ramaiah Nagaraja
- Laboratory of Genetics, National Institute on Aging (NIA), Baltimore, MD, United States of America
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Claudia Schurmann
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Kathrin Budde
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ernst-Moritz-Arndt University Greifswald, Greifswald, Germany
| | - Susan M Farrington
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Evropi Theodoratou
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Antti Jula
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Turku, Finland
| | - Veikko Salomaa
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Cinzia Sala
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | | | - Michel Burnier
- Department of Medicine, Nephrology Division, Lausanne University Hospital, Lausanne, Switzerland
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Norman Klopp
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Sabine Schipf
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Samuli Ripatti
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Turku, Finland; Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom; University of Helsinki, Institute of Molecular Medicine, Helsinki, Finland
| | - Stefano Cabras
- Department of Mathematics and Informatics, Università di Cagliari, Cagliari, Italy; Department of Statistics, Universidad Carlos III de Madrid, Madrid, Spain
| | - Nicole Soranzo
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Teresa Nutile
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nicholas Hastie
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Harry Campbell
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Igor Rudan
- Faculty of Medicine, University of Split, Croatia, Soltanska 2, Split, 21000, Croatia; Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | | | - Chris Haley
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom; Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Oscar H Franco
- Member of Netherlands Consortium for Healthy Aging (NCHA) sponsored by Netherlands Genomics Initiative (NGI), Leiden, The Netherlands; Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Vilmundur Gudnason
- Icelandic Heart Association Research Institute, Kopavogur, Iceland; University of Iceland, Reykjavik, Iceland
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council of Italy, Sassari, Italy
| | - Brenda W Penninx
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands; Department of Epidemiology, Subdivision Genetic Epidemiology, Erasmus MC, Rotterdam, The Netherlands; Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Marina Ciullo
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Peter P Pramstaller
- Center for Biomedicine, European Academy Bozen/Bolzano (EURAC), Bolzano, Italy; Affiliated Institute of the University of Lübeck, Lübeck, Germany
| | - Cornelia M van Duijn
- Department of Epidemiology, Subdivision Genetic Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, United States of America
| | - Giovanni Gambaro
- Institute of Internal Medicine, Renal Program, Columbus-Gemelli University Hospital, Catholic University, Rome, Italy
| | - Ian J Deary
- Department of Psychology, The University of Edinburgh, Edinburgh, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Malcolm G Dunlop
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - James F Wilson
- Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Paolo Gasparini
- Institute for Maternal and Child Health-Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, Italy; University of Trieste, Trieste, Italy
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, 751 85, Sweden
| | - Tim D Spector
- King's College London, St. Thomas' Hospital Campus, London, United Kingdom
| | - Alan F Wright
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| | - Hugh Watkins
- on behalf of PROCARDIS; Department of Cardiovascular Medicine, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Markus Perola
- Estonian Genome Center, University of Tartu, Tartu, Estonia; Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Helsinki, Finland; University of Helsinki, Institute of Molecular Medicine, Helsinki, Finland
| | - Murielle Bochud
- University Institute of Social and Preventive Medicine, Lausanne, Switzerland
| | - W H Linda Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America; Welch Center for Prevention, Epidemiology and Clinical Research, John Hopkins University, Baltimore, MD, United States of America
| | - Mark Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Daniela Toniolo
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milano, Italy
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America; Renal Division, Freiburg University Hospital, Freiburg, Germany
| | - Veronique Vitart
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine (IGMM), University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
269
|
Yu F, Lu J, Liu X, Gazave E, Chang D, Raj S, Hunter-Zinck H, Blekhman R, Arbiza L, Van Hout C, Morrison A, Johnson AD, Bis J, Cupples LA, Psaty BM, Muzny D, Yu J, Gibbs RA, Keinan A, Clark AG, Boerwinkle E. Population genomic analysis of 962 whole genome sequences of humans reveals natural selection in non-coding regions. PLoS One 2015; 10:e0121644. [PMID: 25807536 PMCID: PMC4373932 DOI: 10.1371/journal.pone.0121644] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/14/2014] [Indexed: 12/13/2022] Open
Abstract
Whole genome analysis in large samples from a single population is needed to provide adequate power to assess relative strengths of natural selection across different functional components of the genome. In this study, we analyzed next-generation sequencing data from 962 European Americans, and found that as expected approximately 60% of the top 1% of positive selection signals lie in intergenic regions, 33% in intronic regions, and slightly over 1% in coding regions. Several detailed functional annotation categories in intergenic regions showed statistically significant enrichment in positively selected loci when compared to the null distribution of the genomic span of ENCODE categories. There was a significant enrichment of purifying selection signals detected in enhancers, transcription factor binding sites, microRNAs and target sites, but not on lincRNA or piRNAs, suggesting different evolutionary constraints for these domains. Loci in “repressed or low activity regions” and loci near or overlapping the transcription start site were the most significantly over-represented annotations among the top 1% of signals for positive selection.
Collapse
Affiliation(s)
- Fuli Yu
- Human Genome Sequencing Center, Molecular and Human Genetics Department, Baylor College of Medicine, Houston, Texas, United States of America
- Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- * E-mail: (FY); (EB)
| | - Jian Lu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
- College of Life Sciences, State Key Laboratory of Protein and Plant Gene Research, Center for Bioinformatics, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xiaoming Liu
- Human Genetic Center, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Elodie Gazave
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Diana Chang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Srilakshmi Raj
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Haley Hunter-Zinck
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Ran Blekhman
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Leonardo Arbiza
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Cris Van Hout
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Alanna Morrison
- Human Genetic Center, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Andrew D. Johnson
- National Heart, Lung and Blood Institute (NHLBI) Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Joshua Bis
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, Washington, United States of America
| | - L. Adrienne Cupples
- National Heart, Lung and Blood Institute (NHLBI) Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, Washington, United States of America
| | - Donna Muzny
- Human Genome Sequencing Center, Molecular and Human Genetics Department, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jin Yu
- Human Genome Sequencing Center, Molecular and Human Genetics Department, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Molecular and Human Genetics Department, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alon Keinan
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Andrew G. Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, New York, United States of America
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Molecular and Human Genetics Department, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genetic Center, University of Texas Health Science Center, Houston, Texas, United States of America
- * E-mail: (FY); (EB)
| |
Collapse
|
270
|
Chen L, Zhu W, Mai L, Fang L, Ying K. The association of metabolic syndrome and its components with brachial-ankle pulse wave velocity in south China. Atherosclerosis 2015; 240:345-50. [PMID: 25875386 DOI: 10.1016/j.atherosclerosis.2015.03.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 03/05/2015] [Accepted: 03/18/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Brachial-ankle pulse wave velocity (baPWV) can reflect both central and peripheral arterial stiffness. Metabolic syndrome (MS) and its components may increase arterial stiffness and the risks of cardiovascular diseases. However, the correlation of MS and its components with arterial stiffness has not been not well studied. The aim of this study was to investigate the correlation between MS/its components and arterial stiffness by the measurement of baPWV in south China population. METHODS A total of 8599 subjects were selected from those who underwent health examination in our hospital. MS was defined by Joint Scientific Statement. BaPWV, waist circumference, blood pressure (BP), fasting plasma glucose (FPG), lipid profile and serum uric acid (UA) were measured. The relationship between baPWV and MS/its components was analyzed. RESULTS BaPWV was significantly higher in the subjects with MS than in those without MS (P < 0.001 for both genders). By multivariate regression analysis, all the metabolic components were correlated to baPWV in the male and female subjects except low HDL-C and high UA in the male group. BP and FPG had the strongest correlation factors. The values of baPWV were positively correlated with the advanced age (P < 0.001) and the values of the MS components, and this correlation was stronger in the females than in the males (P < 0.001). CONCLUSION Metabolic syndrome and its individual components were positively correlated with baPWV. Monitoring baPWV is helpful to identify early stage of arterial stiffness in those people with MS.
Collapse
Affiliation(s)
- Liying Chen
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No. 3 Qingchun East Road, Hangzhou 310016, PR China
| | - Wenhua Zhu
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No. 3 Qingchun East Road, Hangzhou 310016, PR China
| | - Linhe Mai
- Shajing People's Hospital, Shenzhen, No. 3 Shajing Street, Baoan District, Shenzhen 518104, PR China
| | - Lizheng Fang
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No. 3 Qingchun East Road, Hangzhou 310016, PR China
| | - Kejing Ying
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No. 3 Qingchun East Road, Hangzhou 310016, PR China.
| |
Collapse
|
271
|
Alonso A, Marsal S, Julià A. Analytical methods in untargeted metabolomics: state of the art in 2015. Front Bioeng Biotechnol 2015; 3:23. [PMID: 25798438 PMCID: PMC4350445 DOI: 10.3389/fbioe.2015.00023] [Citation(s) in RCA: 420] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/18/2015] [Indexed: 12/20/2022] Open
Abstract
Metabolomics comprises the methods and techniques that are used to measure the small molecule composition of biofluids and tissues, and is actually one of the most rapidly evolving research fields. The determination of the metabolomic profile - the metabolome - has multiple applications in many biological sciences, including the developing of new diagnostic tools in medicine. Recent technological advances in nuclear magnetic resonance and mass spectrometry are significantly improving our capacity to obtain more data from each biological sample. Consequently, there is a need for fast and accurate statistical and bioinformatic tools that can deal with the complexity and volume of the data generated in metabolomic studies. In this review, we provide an update of the most commonly used analytical methods in metabolomics, starting from raw data processing and ending with pathway analysis and biomarker identification. Finally, the integration of metabolomic profiles with molecular data from other high-throughput biotechnologies is also reviewed.
Collapse
Affiliation(s)
- Arnald Alonso
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Automatic Control (ESAII), Polytechnic University of Catalonia, Barcelona, Spain
| | - Sara Marsal
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Antonio Julià
- Rheumatology Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| |
Collapse
|
272
|
Takada T, Ichida K, Matsuo H, Nakayama A, Murakami K, Yamanashi Y, Kasuga H, Shinomiya N, Suzuki H. ABCG2 dysfunction increases serum uric acid by decreased intestinal urate excretion. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2015; 33:275-81. [PMID: 24940679 DOI: 10.1080/15257770.2013.854902] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
ATP-binding cassette transporter G2 (ABCG2), also known as breast cancer resistance protein (BCRP), is identified as a high-capacity urate exporter and its dysfunction has an association with serum uric acid (SUA) levels and gout/hyperuricemia risk. However, pathophysiologically important pathway(s) responsible for the ABCG2-mediated urate excretion were unknown. In this study, we investigated how ABCG2 dysfunction affected the urate excretion pathways. First, we revealed that mouse Abcg2 mediates urate transport using the membrane vesicle system. The export process by mouse Abcg2 was ATP-dependent and not saturable under the physiological concentration of urate. Then, we characterized the excretion of urate into urine, bile, and intestinal lumen using in vivo mouse model. SUA of Abcg2-knockout mice was significantly higher than that of control mice. Under this condition, the renal urate excretion was increased in Abcg2-knockout mice, whereas the urate excretion from the intestine was decreased to less than a half. Biliary urate excretion showed no significant difference regardless of Abcg2 genotype. From these results, we estimated the relative contribution of each pathway to total urate excretion; in wild-type mice, the renal excretion pathway contributes approximately two-thirds, the intestinal excretion pathway contributes one-third of the total urate excretion, and the urate excretion into bile is minor. Decreased intestinal excretion could account for the increased SUA of Abcg2-knockout mice. Thus, ABCG2 is suggested to have an important role in extra-renal urate excretion, especially in intestinal excretion. Accordingly, increased SUA in patients with ABCG2 dysfunction could be explained by the decreased excretion of urate from the intestine.
Collapse
Affiliation(s)
- Tappei Takada
- a Department of Pharmacy , The University of Tokyo Hospital , Tokyo , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
273
|
Affiliation(s)
- Asim K. Mandal
- Renal Divisions, Brigham and Women's Hospital and VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts 02115;
| | - David B. Mount
- Renal Divisions, Brigham and Women's Hospital and VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts 02115;
| |
Collapse
|
274
|
Mallamaci F, Testa A, Leonardis D, Tripepi R, Pisano A, Spoto B, Sanguedolce MC, Parlongo RM, Tripepi G, Zoccali C. A Genetic Marker of Uric Acid Level, Carotid Atherosclerosis, and Arterial Stiffness: A Family-Based Study. Am J Kidney Dis 2015; 65:294-302. [DOI: 10.1053/j.ajkd.2014.07.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 07/28/2014] [Indexed: 11/11/2022]
|
275
|
Testa A, Mallamaci F, Leonardis D, Spoto B, Pisano A, Sanguedolce MC, Tripepi G, Zoccali C. Synergism between asymmetric dimethylarginine (ADMA) and a genetic marker of uric acid in CKD progression. Nutr Metab Cardiovasc Dis 2015; 25:167-172. [PMID: 25435339 DOI: 10.1016/j.numecd.2014.10.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/25/2014] [Accepted: 10/27/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND & AIMS We have recently reported that a polymorphism (rs734553) in a major urate transporter gene (GLUT9) is a strong predictor of incident renal events in stage 2-5 CKD patients implying that life-time exposure to high uric acid levels may be causally implicated in CKD progression. Since disturbed NO bioavailability is a major pathway whereby high uric may cause renal damage, we tested the interaction between the major endogenous inhibitor of NO synthase, asymmetric-dimethylargine (ADMA), and the rs734553 polymorphism for CKD progression in the same cohort. METHODS & RESULTS Over a 29 ± 11 months follow-up the risk for incident renal events was higher in patients harboring the risk allele of the polymorphism (T) as compared to those without the risk allele (HR: 2.35, 95% CI: 1.25-4.42, P = 0.008) (p = 0.01). Similarly, patients with ADMA > median value had an increased risk for the same outcome (HR: 1.37, 95% CI: 1.06-1.76, P = 0.016). Interaction analysis showed a strong amplification by ADMA of the risk for renal events associated to the T allele because in adjusted (P = 0.016) and bootstrapping validated (P = 0.020) analyses the risk excess associated to this allele was progressively higher across increasing ADMA levels. CONCLUSIONS The rs734553 polymorphism, the strongest genetic marker of uric acid levels discovered so far, interacts with ADMA in determining the risk for CKD progression in CKD patients. This synergic interaction conforms to biological knowledge indicating that disturbed NO bio-availability is a critical pathway whereby life time exposure to high uric acid may engender renal damage.
Collapse
Affiliation(s)
- A Testa
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy
| | - F Mallamaci
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy
| | - D Leonardis
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy
| | - B Spoto
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy
| | - A Pisano
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy
| | - M C Sanguedolce
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy
| | - G Tripepi
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy
| | - C Zoccali
- CNR-IFC/IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Ospedali Riuniti, 89124 Reggio Calabria, Italy.
| |
Collapse
|
276
|
Kuo CF, Grainge MJ, See LC, Yu KH, Luo SF, Valdes AM, Zhang W, Doherty M. Familial aggregation of gout and relative genetic and environmental contributions: a nationwide population study in Taiwan. Ann Rheum Dis 2015; 74:369-74. [PMID: 24265412 PMCID: PMC4316854 DOI: 10.1136/annrheumdis-2013-204067] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/22/2013] [Accepted: 11/03/2013] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To examine familial aggregation of gout and to estimate the heritability and environmental contributions to gout susceptibility in the general population. METHODS Using data from the National Health Insurance (NHI) Research Database in Taiwan, we conducted a nationwide cross-sectional study of data collected from 22 643 748 beneficiaries of the NHI in 2004; among them 1 045 059 individuals had physician-diagnosed gout. We estimated relative risks (RR) of gout in individuals with affected first-degree and second-degree relatives and relative contributions of genes (heritability), common environment shared by family members and non-shared environment to gout susceptibility. RESULTS RRs for gout were significantly higher in individuals with affected first-degree relatives (men, 1.91 (95% CI 1.90 to 1.93); women, 1.97 (95% CI 1.94 to 1.99)) and also in those with affected second-degree relatives (men, 1.27 (95% CI 1.23 to 1.31); women, 1.40 (95% CI 1.35 to 1.46)). RRs (95% CIs) for individuals with an affected twin, sibling, offspring, parent, grandchild, nephew/niece, uncle/aunt and grandparent were 8.02 (6.95 to 9.26), 2.59 (2.54 to 2.63), 1.96 (1.95 to 1.97), 1.93 (1.91 to 1.94), 1.48 (1.43 to 1.53), 1.40 (1.32 to 1.47), 1.31 (1.24 to 1.39), and 1.26 (1.21 to 1.30), respectively. The relative contributions of heritability, common and non-shared environmental factors to phenotypic variance of gout were 35.1, 28.1 and 36.8% in men and 17.0, 18.5 and 64.5% in women, respectively. CONCLUSIONS This population-based study confirms that gout aggregates within families. The risk of gout is higher in people with a family history. Genetic and environmental factors contribute to gout aetiology, and the relative contributions are sexually dimorphic.
Collapse
Affiliation(s)
- Chang-Fu Kuo
- Department of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Matthew J Grainge
- Division of Epidemiology and Public Health, School of Medicine, University of Nottingham, Nottingham, UK
| | - Lai-Chu See
- Department of Public Health, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Biostatistics Core Laboratory, Molecular Medicine Research Centre, Chang Gung University, Taoyuan, Taiwan
| | - Kuang-Hui Yu
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shue-Fen Luo
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ana M Valdes
- Department of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Weiya Zhang
- Department of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Michael Doherty
- Department of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
277
|
Han X, Gui L, Liu B, Wang J, Li Y, Dai X, Li J, Yang B, Qiu G, Feng J, Zhang X, Wu T, He M. Associations of the uric acid related genetic variants in SLC2A9 and ABCG2 loci with coronary heart disease risk. BMC Genet 2015; 16:4. [PMID: 25634581 PMCID: PMC4314773 DOI: 10.1186/s12863-015-0162-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 01/05/2015] [Indexed: 12/01/2022] Open
Abstract
Background Multiple studies investigated the associations between serum uric acid and coronary heart disease (CHD) risk. However, further investigations still remain to be carried out to determine whether there exists a causal relationship between them. We aim to explore the associations between genetic variants in uric acid related loci of SLC2A9 and ABCG2 and CHD risk in a Chinese population. Results A case–control study including 1,146 CHD cases and 1,146 controls was conducted. Association analysis between two uric acid related variants (SNP rs11722228 in SLC2A9 and rs4148152 in ABCG2) and CHD risk was performed by logistic regression model. Adjusted odds ratios (ORs) with 95% confidence intervals (CIs) were calculated. Compared with subjects with A allele of rs4148152, those with G allele had a decreased CHD risk and the association remained significant in a multivariate model. However, it altered to null when BMI was added into the model. No significant association was observed between rs11722228 and CHD risk. The distribution of CHD risk factors was not significantly different among different genotypes of both SNPs. Among subjects who did not consume alcohol, the G allele of rs4148152 showed a moderate protective effect. However, no significant interactions were observed between SNP by CHD risk factors on CHD risk. Conclusions There might be no association between the two uric acid related SNPs with CHD risk. Further studies were warranted to validate these results. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0162-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xu Han
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Lixuan Gui
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Bing Liu
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Jing Wang
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Yaru Li
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiayun Dai
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Li
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Binyao Yang
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaokun Qiu
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Jing Feng
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaomin Zhang
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Tangchun Wu
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China.
| | - Meian He
- Institute of Occupational Medicine and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, China. .,MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, 13 Hangkong Rd, Wuhan, Hubei, 430030, China.
| |
Collapse
|
278
|
Common variants related to serum uric acid concentrations are associated with glucose metabolism and insulin secretion in a Chinese population. PLoS One 2015; 10:e0116714. [PMID: 25617895 PMCID: PMC4305305 DOI: 10.1371/journal.pone.0116714] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 12/13/2014] [Indexed: 01/11/2023] Open
Abstract
Background Elevated serum uric acid concentration is an independent risk factor and predictor of type 2 diabetes (T2D). Whether the uric acid-associated genes have an impact on T2D remains unclear. We aimed to investigate the effects of the uric acid-associated genes on the risk of T2D as well as glucose metabolism and insulin secretion. Method We recruited 2,199 normal glucose tolerance subjects from the Shanghai Diabetes Study I and II and 2,999 T2D patients from the inpatient database of Shanghai Diabetes Institute. Fifteen single nucleotide polymorphisms (SNPs) mapped in or near 11 loci (PDZK1, GCKR, LRP2, SLC2A9, ABCG2, LRRC16A, SLC17A1, SLC17A3, SLC22A11, SLC22A12 and SF1) were genotyped and serum biochemical parameters related to uric acid and T2D were determined. Results SF1 rs606458 showed strong association to T2D in both males and females (p = 0.034 and 0.0008). In the males, LRRC16A was associated with 2-h insulin and insulin secretion (p = 0.009 and 0.009). SLC22A11 was correlated with HOMA-B and insulin secretion (p = 0.048 and 0.029). SLC2A9 rs3775948 was associated with 2-h glucose (p = 0.043). In the females, LRP2 rs2544390 and rs1333049 showed correlations with fasting insulin, HOMA-IR and insulin secretion (p = 0.028, 0.033 and 0.052 and p = 0.034, 0.047 and 0.038, respectively). SLC2A9 rs11722228 was correlated with 2-h glucose, 2-h insulin and insulin secretion (p = 0.024, 0.049 and 0.049, respectively). Conclusions Our results indicated that the uric acid-associated genes have an impact on the risk of T2D, glucose metabolism and insulin secretion in a Chinese population.
Collapse
|
279
|
Jin TB, Ren Y, Shi X, Jiri M, He N, Feng T, Yuan D, Kang L. Genetic variations in the CLNK gene and ZNF518B gene are associated with gout in case-control sample sets. Rheumatol Int 2015; 35:1141-7. [PMID: 25591661 DOI: 10.1007/s00296-015-3215-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/08/2015] [Indexed: 12/19/2022]
Abstract
A genome-wide association study of gout in European populations identified 12 genetic variants strongly associated with risk of gout, but it is unknown whether these variants are also associated with gout risk in Chinese populations. A total of 145 patients with gout and 310 healthy control patients were recruited for a case-control association study. Twelve SNPs of CLNK and ZNF518B gene were genotyped, and association analysis was performed. Odds ratios (ORs) with 95 % confidence intervals (CIs) were used to assess the association. Overall, we found four risk alleles for gout in patients: the allele "G" of rs2041215 and rs1686947 in the CLNK gene by dominant model (OR 1.66; 95 % CI 1.04-2.63; p = 0.031) (OR 2.19; 95 % CI 1.38-3.46; p = 0.001) and additive model (OR 1.39; 95 % CI 1.00-1.93; p = 0.049) (OR 1.67; 95 % CI 1.19-2.32; p = 0.003), respectively, and the allele "A" of rs10938799 and rs10016022 in ZNF518B gene by recessive model (OR 4.66; 95 % CI 1.44-15.09; p = 0.008) (OR 4.54; 95 % CI 1.23-16.76; p = 0.020). Further haplotype analysis showed that the TCATTCTGA haplotype of CLNK was more frequent among patients with gout (adjusted OR 0.48; 95 % CI 0.24-0.95; p = 0.036). Additionally, polymorphisms of rs2041215, rs10938799, and rs17467273 were also correlated with clinical pathological parameters. This study provides evidence for gout susceptibility genes, CLNK and ZNF518B, in a Chinese population, which may have potential as diagnostic and prognostic marker for gout patients.
Collapse
Affiliation(s)
- Tian-Bo Jin
- Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Tibet University for Nationalities, Xianyang, 712082, Shaanxi, China
| | | | | | | | | | | | | | | |
Collapse
|
280
|
Demirkan A, Henneman P, Verhoeven A, Dharuri H, Amin N, van Klinken JB, Karssen LC, de Vries B, Meissner A, Göraler S, van den Maagdenberg AMJM, Deelder AM, C ’t Hoen PA, van Duijn CM, van Dijk KW. Insight in genome-wide association of metabolite quantitative traits by exome sequence analyses. PLoS Genet 2015; 11:e1004835. [PMID: 25569235 PMCID: PMC4287344 DOI: 10.1371/journal.pgen.1004835] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 10/16/2014] [Indexed: 12/20/2022] Open
Abstract
Metabolite quantitative traits carry great promise for epidemiological studies, and their genetic background has been addressed using Genome-Wide Association Studies (GWAS). Thus far, the role of less common variants has not been exhaustively studied. Here, we set out a GWAS for metabolite quantitative traits in serum, followed by exome sequence analysis to zoom in on putative causal variants in the associated genes. 1H Nuclear Magnetic Resonance (1H-NMR) spectroscopy experiments yielded successful quantification of 42 unique metabolites in 2,482 individuals from The Erasmus Rucphen Family (ERF) study. Heritability of metabolites were estimated by SOLAR. GWAS was performed by linear mixed models, using HapMap imputations. Based on physical vicinity and pathway analyses, candidate genes were screened for coding region variation using exome sequence data. Heritability estimates for metabolites ranged between 10% and 52%. GWAS replicated three known loci in the metabolome wide significance: CPS1 with glycine (P-value = 1.27×10−32), PRODH with proline (P-value = 1.11×10−19), SLC16A9 with carnitine level (P-value = 4.81×10−14) and uncovered a novel association between DMGDH and dimethyl-glycine (P-value = 1.65×10−19) level. In addition, we found three novel, suggestively significant loci: TNP1 with pyruvate (P-value = 1.26×10−8), KCNJ16 with 3-hydroxybutyrate (P-value = 1.65×10−8) and 2p12 locus with valine (P-value = 3.49×10−8). Exome sequence analysis identified potentially causal coding and regulatory variants located in the genes CPS1, KCNJ2 and PRODH, and revealed allelic heterogeneity for CPS1 and PRODH. Combined GWAS and exome analyses of metabolites detected by high-resolution 1H-NMR is a robust approach to uncover metabolite quantitative trait loci (mQTL), and the likely causative variants in these loci. It is anticipated that insight in the genetics of intermediate phenotypes will provide additional insight into the genetics of complex traits. Human metabolic individuality is under strict control of genetic and environmental factors. In our study, we aimed to find the genetic determinants of circulating molecules in sera of large set of individuals representing the general population. First, we performed a hypothesis-free genome wide screen in this population to identify genetic regions of interest. Our study confirmed four known gene metabolite connections, but also pointed to four novel ones. Genome-wide screens enriched for common intergenic variants may miss causal genetic variations directly changing the protein sequence. To investigate this further, we zoomed into regions of interest and tested whether the association signals obtained in the first stage were direct, or whether they represent causal variations, which were not captured in the initial panel. These subsequent tests showed that protein coding and regulatory variations are involved in metabolite levels. For two genomic regions we also found that genes harbour more than one causal variant influencing metabolite levels independent of each other. We also observed strong connection between markers of cardio-metabolic health and metabolites. Taken together, our novel loci are of interest for further research to investigate the causal relation to for instance type 2 diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Ayşe Demirkan
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Peter Henneman
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Harish Dharuri
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jan Bert van Klinken
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lennart C. Karssen
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Boukje de Vries
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Axel Meissner
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sibel Göraler
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Arn M. J. M. van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - André M. Deelder
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter A. C ’t Hoen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
- * E-mail:
| |
Collapse
|
281
|
Hegedüs C, Hegedüs T, Sarkadi B. The Role of ABC Multidrug Transporters in Resistance to Targeted Anticancer Kinase Inhibitors. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2015. [DOI: 10.1007/978-3-319-09801-2_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
282
|
Krumsiek J, Mittelstrass K, Do KT, Stückler F, Ried J, Adamski J, Peters A, Illig T, Kronenberg F, Friedrich N, Nauck M, Pietzner M, Mook-Kanamori DO, Suhre K, Gieger C, Grallert H, Theis FJ, Kastenmüller G. Gender-specific pathway differences in the human serum metabolome. Metabolomics 2015; 11:1815-1833. [PMID: 26491425 PMCID: PMC4605991 DOI: 10.1007/s11306-015-0829-0] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/08/2015] [Indexed: 02/07/2023]
Abstract
The susceptibility for various diseases as well as the response to treatments differ considerably between men and women. As a basis for a gender-specific personalized healthcare, an extensive characterization of the molecular differences between the two genders is required. In the present study, we conducted a large-scale metabolomics analysis of 507 metabolic markers measured in serum of 1756 participants from the German KORA F4 study (903 females and 853 males). One-third of the metabolites show significant differences between males and females. A pathway analysis revealed strong differences in steroid metabolism, fatty acids and further lipids, a large fraction of amino acids, oxidative phosphorylation, purine metabolism and gamma-glutamyl dipeptides. We then extended this analysis by a network-based clustering approach. Metabolite interactions were estimated using Gaussian graphical models to get an unbiased, fully data-driven metabolic network representation. This approach is not limited to possibly arbitrary pathway boundaries and can even include poorly or uncharacterized metabolites. The network analysis revealed several strongly gender-regulated submodules across different pathways. Finally, a gender-stratified genome-wide association study was performed to determine whether the observed gender differences are caused by dimorphisms in the effects of genetic polymorphisms on the metabolome. With only a single genome-wide significant hit, our results suggest that this scenario is not the case. In summary, we report an extensive characterization and interpretation of gender-specific differences of the human serum metabolome, providing a broad basis for future analyses.
Collapse
Affiliation(s)
- Jan Krumsiek
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Kirstin Mittelstrass
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kieu Trinh Do
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ferdinand Stückler
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Janina Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jerzy Adamski
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Neuherberg, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Cardiovascular Disease Research (DZHK e.V.), Munich, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Greifswald, Greifswald, Germany
| | - Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Greifswald, Greifswald, Germany
| | - Dennis O. Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Endocrinology, Leiden University Medical Center, Leiden, Netherlands
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Qatar Foundation, Doha, Qatar
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Qatar Foundation, Doha, Qatar
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Mathematics, Technische Universität München, Garching, Germany
| | - Gabi Kastenmüller
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
283
|
Kurajoh M, Koyama H, Hatayama M, Okazaki H, Shoji T, Moriwaki Y, Yamamoto T, Nakayama T, Namba M. Partial HPRT Deficiency with a Novel Mutation of the HPRT Gene in Combination with Four Previously Reported Variants Associated with Hyperuricemia. Intern Med 2015; 54:1523-6. [PMID: 26073243 DOI: 10.2169/internalmedicine.54.3290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A 15-year-old boy was referred to our department due to gout. The laboratory findings showed hyperuricemia with a decreased erythrocyte hypoxanthine phosphoribosyl transferase (HPRT) activity. The HPRT cDNA sequence was revealed to be 206A>T, which has not been previously reported. In addition, direct sequencing of genomic DNA showed the patient to possess four variants reported to be associated with hyperuricemia. This is the first case report of partial HPRT deficiency due to a novel HPRT mutation accompanied by variants associated with hyperuricemia. Combination treatment consisting of benzbromarone and febuxostat had a significant effect in reducing the urate level in our patient.
Collapse
Affiliation(s)
- Masafumi Kurajoh
- Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
284
|
Yang H, Gao L, Niu Y, Zhou Y, Lin H, Jiang J, Kong X, Liu X, Li L. Mangiferin Inhibits Renal Urate Reabsorption by Modulating Urate Transporters in Experimental Hyperuricemia. Biol Pharm Bull 2015; 38:1591-8. [DOI: 10.1248/bpb.b15-00402] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hua Yang
- Biomedical Engineering Research Center, Kunming Medical University
- The Second Affiliated Hospital of Kunming Medical University
| | - Lihui Gao
- Biomedical Engineering Research Center, Kunming Medical University
| | - Yanfen Niu
- Biomedical Engineering Research Center, Kunming Medical University
| | - Yuanfang Zhou
- Biomedical Engineering Research Center, Kunming Medical University
| | - Hua Lin
- Biomedical Engineering Research Center, Kunming Medical University
| | | | | | - Xu Liu
- Biomedical Engineering Research Center, Kunming Medical University
| | - Ling Li
- Biomedical Engineering Research Center, Kunming Medical University
| |
Collapse
|
285
|
Etiology and pathogenesis of gout. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00187-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
286
|
Nigam SK, Bush KT, Martovetsky G, Ahn SY, Liu HC, Richard E, Bhatnagar V, Wu W. The organic anion transporter (OAT) family: a systems biology perspective. Physiol Rev 2015; 95:83-123. [PMID: 25540139 PMCID: PMC4281586 DOI: 10.1152/physrev.00025.2013] [Citation(s) in RCA: 349] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The organic anion transporter (OAT) subfamily, which constitutes roughly half of the SLC22 (solute carrier 22) transporter family, has received a great deal of attention because of its role in handling of common drugs (antibiotics, antivirals, diuretics, nonsteroidal anti-inflammatory drugs), toxins (mercury, aristolochic acid), and nutrients (vitamins, flavonoids). Oats are expressed in many tissues, including kidney, liver, choroid plexus, olfactory mucosa, brain, retina, and placenta. Recent metabolomics and microarray data from Oat1 [Slc22a6, originally identified as NKT (novel kidney transporter)] and Oat3 (Slc22a8) knockouts, as well as systems biology studies, indicate that this pathway plays a central role in the metabolism and handling of gut microbiome metabolites as well as putative uremic toxins of kidney disease. Nuclear receptors and other transcription factors, such as Hnf4α and Hnf1α, appear to regulate the expression of certain Oats in conjunction with phase I and phase II drug metabolizing enzymes. Some Oats have a strong selectivity for particular signaling molecules, including cyclic nucleotides, conjugated sex steroids, odorants, uric acid, and prostaglandins and/or their metabolites. According to the "Remote Sensing and Signaling Hypothesis," which is elaborated in detail here, Oats may function in remote interorgan communication by regulating levels of signaling molecules and key metabolites in tissues and body fluids. Oats may also play a major role in interorganismal communication (via movement of small molecules across the intestine, placental barrier, into breast milk, and volatile odorants into the urine). The role of various Oat isoforms in systems physiology appears quite complex, and their ramifications are discussed in the context of remote sensing and signaling.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Kevin T Bush
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Gleb Martovetsky
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Sun-Young Ahn
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Henry C Liu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Erin Richard
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Vibha Bhatnagar
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| | - Wei Wu
- Departments of Pediatrics, Medicine, Cellular and Molecular Medicine, Bioengineering, and Family and Preventative Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
287
|
Sull JW, Yang SJ, Kim S, Jee SH. The ABCG2 Polymorphism rs2725220 Is Associated with Hyperuricemia in the Korean Population. Genomics Inform 2014; 12:231-5. [PMID: 25705163 PMCID: PMC4330259 DOI: 10.5808/gi.2014.12.4.231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/06/2014] [Accepted: 11/06/2014] [Indexed: 12/24/2022] Open
Abstract
Elevated serum uric acid levels are associated with a variety of adverse health outcomes, including gout, hypertension, diabetes mellitus, metabolic syndrome, and cardiovascular diseases. Several genome-wide association studies on uric acid levels have implicated the ATP-binding cassette, subfamily G, member 2 (ABCG2) gene as being possibly causal. We investigated an association between the single-nucleotide polymorphism (SNP) rs2725220 in the ABCG2 gene and uric acid levels in the Korean population. A total of 991 subjects in Seoul City were used for a replication study with ABCG2 SNP rs2725220. The rs2725220 SNP in the ABCG2 gene was associated with mean uric acid levels (effect per allele 0.25 mg/dL, p < 0.0001). Subjects with the GC/CC genotype had a 1.78-fold (range, 1.22- to 2.62-fold) higher risk of having abnormal uric acid levels (≥7.0 mg/dL) than subjects with the GG genotype. When analyzed by gender, the association with ABCG2 was stronger in men than in women. The association with ABCG2 was much stronger in male subjects with body mass index (BMI) ≥ 26.4 (odds ratio, 5.09; 95% confidence interval, 2.41 to 10.8) than in male subjects with BMI < 26.4. This study clearly demonstrates that genetic variations in ABCG2 influence uric acid levels in Korean adults.
Collapse
Affiliation(s)
- Jae Woong Sull
- Department of Biomedical Laboratory Science, College of Health Sciences, Eulji University, Seongnam 461-713, Korea
| | - Seung-Ju Yang
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 302-718, Korea
| | - Soriul Kim
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul 120-752, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul 120-752, Korea
| |
Collapse
|
288
|
Dong Z, Guo S, Yang Y, Wu J, Guan M, Zou H, Jin L, Wang J. Association between ABCG2 Q141K polymorphism and gout risk affected by ethnicity and gender: a systematic review and meta-analysis. Int J Rheum Dis 2014; 18:382-91. [PMID: 25639607 DOI: 10.1111/1756-185x.12519] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIM Original studies have employed various genetic models in association analysis between ABCG2 Q141K (rs2231142) with gout risk and different or conflicting results, especially regarding the role of gender in this association. In addition, it is not clear whether the association varies by ethnicity. METHOD Articles published before September 1, 2013 were extracted and registered into databases for the systematic review of this polymorphism. The quality of each study was scored based on predefined criteria. The genetic model was identified through stratification analysis, then a meta-analysis including all publically available data was preformed to test the association between rs2231142 and gout risk. Potential sources of heterogeneity were sought out via stratification analysis and meta-regression analysis. RESULTS Nine case-control studies involving 17 942 individuals were eligible for the meta-analysis of rs2231142. Codominant model was the most appropriate genetic model to interpret the susceptibility cause. It showed that the rs2231142 T allele obviously increased gout risk, and TT was much stronger than GT (TT vs. GG: OR, 4.10; 95% CI, 2.90-5.80; GT vs. GG: OR, 1.71, 95% CI, 1.39-2.10). In addition, gender and ethnicity were found to affect the association between the susceptibility of gout and rs2231142. CONCLUSION ABCG2 rs2231142 is an important genetic factor in increasing gout risk, and the difference in genetic association has been found between male and female populations. In addition, the degree of association has been found to vary with ethnicity.
Collapse
Affiliation(s)
- Zheng Dong
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Shicheng Guo
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yajun Yang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Junjie Wu
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Ming Guan
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Hejian Zou
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.,Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Jin
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| |
Collapse
|
289
|
Abstract
Potential drug-drug interactions mediated by the ATP-binding cassette (ABC) transporter and solute carrier (SLC) transporter families are of clinical and regulatory concern. However, the endogenous functions of these drug transporters are not well understood. Discussed here is evidence for the roles of ABC and SLC transporters in the handling of diverse substrates, including metabolites, antioxidants, signalling molecules, hormones, nutrients and neurotransmitters. It is suggested that these transporters may be part of a larger system of remote communication ('remote sensing and signalling') between cells, organs, body fluid compartments and perhaps even separate organisms. This broader view may help to clarify disease mechanisms, drug-metabolite interactions and drug effects relevant to diabetes, chronic kidney disease, metabolic syndrome, hypertension, gout, liver disease, neuropsychiatric disorders, inflammatory syndromes and organ injury, as well as prenatal and postnatal development.
Collapse
Affiliation(s)
- Sanjay K Nigam
- Departments of Pediatrics, Medicine, and Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0693, USA
| |
Collapse
|
290
|
Hall MA, Verma A, Brown-Gentry KD, Goodloe R, Boston J, Wilson S, McClellan B, Sutcliffe C, Dilks HH, Gillani NB, Jin H, Mayo P, Allen M, Schnetz-Boutaud N, Crawford DC, Ritchie MD, Pendergrass SA. Detection of pleiotropy through a Phenome-wide association study (PheWAS) of epidemiologic data as part of the Environmental Architecture for Genes Linked to Environment (EAGLE) study. PLoS Genet 2014; 10:e1004678. [PMID: 25474351 PMCID: PMC4256091 DOI: 10.1371/journal.pgen.1004678] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/16/2014] [Indexed: 12/19/2022] Open
Abstract
We performed a Phenome-wide association study (PheWAS) utilizing diverse genotypic and phenotypic data existing across multiple populations in the National Health and Nutrition Examination Surveys (NHANES), conducted by the Centers for Disease Control and Prevention (CDC), and accessed by the Epidemiological Architecture for Genes Linked to Environment (EAGLE) study. We calculated comprehensive tests of association in Genetic NHANES using 80 SNPs and 1,008 phenotypes (grouped into 184 phenotype classes), stratified by race-ethnicity. Genetic NHANES includes three surveys (NHANES III, 1999-2000, and 2001-2002) and three race-ethnicities: non-Hispanic whites (n = 6,634), non-Hispanic blacks (n = 3,458), and Mexican Americans (n = 3,950). We identified 69 PheWAS associations replicating across surveys for the same SNP, phenotype-class, direction of effect, and race-ethnicity at p<0.01, allele frequency >0.01, and sample size >200. Of these 69 PheWAS associations, 39 replicated previously reported SNP-phenotype associations, 9 were related to previously reported associations, and 21 were novel associations. Fourteen results had the same direction of effect across more than one race-ethnicity: one result was novel, 11 replicated previously reported associations, and two were related to previously reported results. Thirteen SNPs showed evidence of pleiotropy. We further explored results with gene-based biological networks, contrasting the direction of effect for pleiotropic associations across phenotypes. One PheWAS result was ABCG2 missense SNP rs2231142, associated with uric acid levels in both non-Hispanic whites and Mexican Americans, protoporphyrin levels in non-Hispanic whites and Mexican Americans, and blood pressure levels in Mexican Americans. Another example was SNP rs1800588 near LIPC, significantly associated with the novel phenotypes of folate levels (Mexican Americans), vitamin E levels (non-Hispanic whites) and triglyceride levels (non-Hispanic whites), and replication for cholesterol levels. The results of this PheWAS show the utility of this approach for exposing more of the complex genetic architecture underlying multiple traits, through generating novel hypotheses for future research.
Collapse
Affiliation(s)
- Molly A. Hall
- Center for Systems Genomics, Department of Biochemistry and Molecular Biology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Anurag Verma
- Center for Systems Genomics, Department of Biochemistry and Molecular Biology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Kristin D. Brown-Gentry
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Robert Goodloe
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jonathan Boston
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Sarah Wilson
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Bob McClellan
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Cara Sutcliffe
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Holly H. Dilks
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Nila B. Gillani
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Hailing Jin
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ping Mayo
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Melissa Allen
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Nathalie Schnetz-Boutaud
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Dana C. Crawford
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Marylyn D. Ritchie
- Center for Systems Genomics, Department of Biochemistry and Molecular Biology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Sarah A. Pendergrass
- Center for Systems Genomics, Department of Biochemistry and Molecular Biology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
291
|
Serum uric acid levels are associated with polymorphisms in the SLC2A9, SF1, and GCKR genes in a Chinese population. Acta Pharmacol Sin 2014; 35:1421-7. [PMID: 25283508 DOI: 10.1038/aps.2014.87] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/31/2014] [Indexed: 01/21/2023]
Abstract
AIM Genome-wide association studies have identified several novel loci associated with serum uric acid concentrations in individuals of European descent. In the current study, we aimed to evaluate the associations between these loci and serum uric acid concentrations in a Chinese population. METHODS Fourteen single nucleotide polymorphisms (SNPs) mapped in or near 11 loci (PDZK1, GCKR, LRP2, SLC2A9, ABCG2, LRRC16A, SLC17A1, SLC17A3, SLC22A11, SLC22A12 and SF1) were genotyped in 2329 Chinese subjects in Shanghai. Serum biochemical parameters including uric acid concentrations were determined. All the variants were analyzed for gender differences since uric acid metabolism differed between genders. RESULTS In males after adjustments for age and BMI, GCKR rs780094, SLC2A9 rs11722228 and SF1 rs606458 were associated with the uric acid concentrations, which were statistically significant (P=0.016, 0.001 and 0.03, respectively), whereas SLC2A9 rs3775948 was marginally associated with the uric acid concentrations (P=0.071). In females, SLC22A12 rs506338 was also marginally associated with the uric acid concentrations (P=0.057). The meta-analysis for combined data from both males and females revealed that rs3775948 and rs606458 were associated with the uric acid concentrations (P=0.036 and 0.043, respectively). Furthermore, the gender significantly affected the association of rs11722228 with serum uric acid levels (P=0.012). CONCLUSION The SLC2A9 rs11722228, SF1 rs606458 and GCKR rs780094 variants modulate uric acid concentrations in Chinese males, while SF1 rs606458 and SLC2A9 rs3775948 are associated with the uric acid concentrations in both Chinese males and females.
Collapse
|
292
|
Vandell AG, McDonough CW, Gong Y, Langaee TY, Lucas AM, Chapman AB, Gums JG, Beitelshees AL, Bailey KR, Johnson RJ, Boerwinkle E, Turner ST, Cooper-DeHoff RM, Johnson JA. Hydrochlorothiazide-induced hyperuricaemia in the pharmacogenomic evaluation of antihypertensive responses study. J Intern Med 2014; 276:486-97. [PMID: 24612202 PMCID: PMC4130802 DOI: 10.1111/joim.12215] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Elevations in uric acid (UA) and the associated hyperuricaemia are commonly observed secondary to treatment with thiazide diuretics. We sought to identify novel single nucleotide polymorphisms (SNPs) associated with hydrochlorothiazide (HCTZ)-induced elevations in UA and hyperuricaemia. METHODS A genome-wide association study of HCTZ-induced changes in UA was performed in Caucasian and African American participants from the pharmacogenomic evaluation of antihypertensive responses (PEAR) study who were treated with HCTZ monotherapy. Suggestive SNPs were replicated in Caucasians and African Americans from the PEAR study who were treated with HCTZ add-on therapy. Replicated regions were followed up through expression and pathway analysis. RESULTS Five unique gene regions were identified in African Americans (LUC7L2, ANKRD17/COX18, FTO, PADI4 and PARD3B), and one region was identified in Caucasians (GRIN3A). Increases in UA of up to 1.8 mg dL(-1) were observed following HCTZ therapy in individuals homozygous for risk alleles, with heterozygotes displaying an intermediate phenotype. Several risk alleles were also associated with an increased risk of HCTZ-induced clinical hyperuricaemia. A composite risk score, constructed in African Americans using the 'top' SNP from each gene region, was strongly associated with HCTZ-induced UA elevations (P = 1.79 × 10(-7) ) and explained 11% of the variability in UA response. Expression studies in RNA from whole blood revealed significant differences in expression of FTO by rs4784333 genotype. Pathway analysis showed putative connections between many of the genes identified through common microRNAs. CONCLUSION Several novel gene regions were associated with HCTZ-induced UA elevations in African Americans (LUC7L2, COX18/ANKRD17, FTO, PADI4 and PARD3B), and one region was associated with these elevations in Caucasians (GRIN3A).
Collapse
Affiliation(s)
- A G Vandell
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
293
|
Abstract
Gout is a common inflammatory arthritis triggered by the crystallization of uric acid within the joints. Gout affects millions worldwide and has an increasing prevalence. Recent research has been carried out to better qualify and quantify the risk factors predisposing individuals to gout. These can largely be broken into nonmodifiable risk factors, such as gender, age, race, and genetics, and modifiable risk factors, such as diet and lifestyle. Increasing knowledge of factors predisposing certain individuals to gout could potentially lead to improved preventive practices. This review summarizes the nonmodifiable and modifiable risk factors associated with development of gout.
Collapse
Affiliation(s)
- Lindsey A MacFarlane
- Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Seoyoung C Kim
- Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women's Hospital, 1620 Tremont Street, Suite 3030, Boston, MA 02120, USA; Division of Rheumatology, Allergy and Immunology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
294
|
Hale C, Lloyd DJ, Pellacani A, Véniant MM. Molecular targeting of the GK-GKRP pathway in diabetes. Expert Opin Ther Targets 2014; 19:129-39. [DOI: 10.1517/14728222.2014.965681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
295
|
Simon KC, Eberly S, Gao X, Oakes D, Tanner CM, Shoulson I, Fahn S, Schwarzschild MA, Ascherio A. Mendelian randomization of serum urate and parkinson disease progression. Ann Neurol 2014; 76:862-8. [PMID: 25257975 DOI: 10.1002/ana.24281] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Higher serum urate concentrations predict more favorable prognosis in individuals with Parkinson disease (PD). The purpose of this study was to test the causality of this association using a Mendelian randomization approach. METHODS The study was conducted among participants in DATATOP and PRECEPT, 2 randomized trials among patients with early PD. The 808 patients with available DNA were genotyped for 3 SLC2A9 single nucleotide polymorphisms (SNPs) that identify an allele associated with lower urate concentrations, and for selected SNPs in other genes encoding urate transporters that have modest or no effect on serum urate levels. An SLC2A9 score was created based on the total number of minor alleles at the 3 SLC2A9 loci. Primary outcome was disability requiring dopaminergic treatment. RESULTS Serum urate concentrations were 0.69mg/dl lower among individuals with ≥4 SLC2A9 minor alleles as compared to those with ≤2 (p = 0.0002). The hazard ratio (HR) for progression to disability requiring dopaminergic treatment increased with increasing SLC2A9 score (HR = 1.16, 95% confidence interval [CI] = 1.00-1.35, p = 0.056). In a comparative analysis, the HR was 1.27 (95% CI = 1.00-1.61, p = 0.0497) for a 0.5mg/dl genetically conferred decrease in serum urate, and 1.05 (95% CI = 1.01-1.10, p = 0.0133) for a 0.5mg/dl decrease in measured serum urate. No associations were found between polymorphisms in other genes associated with urate that do not affect serum urate and PD progression. INTERPRETATION This Mendelian randomization analysis adds to the evidence of a causal protective effect of high urate levels.
Collapse
Affiliation(s)
- Kelly Claire Simon
- Department of Nutrition, Harvard School of Public Health, Boston, MA; Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
296
|
Gilks WP, Abbott JK, Morrow EH. Sex differences in disease genetics: evidence, evolution, and detection. Trends Genet 2014; 30:453-63. [DOI: 10.1016/j.tig.2014.08.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 12/13/2022]
|
297
|
Jansen L, de Niet A, Stelma F, van Iperen EPA, van Dort KA, Plat-Sinnige MJT, Takkenberg RB, Chin DJ, Zwinderman AHK, Lopatin U, Kootstra NA, Reesink HW. HBsAg loss in patients treated with peginterferon alfa-2a and adefovir is associated with SLC16A9 gene variation and lower plasma carnitine levels. J Hepatol 2014; 61:730-7. [PMID: 24824278 DOI: 10.1016/j.jhep.2014.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 04/16/2014] [Accepted: 05/05/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Achievement of HBsAg loss remains the hallmark of chronic hepatitis B treatment. In order to identify host factors contributing to treatment-induced HBsAg loss, we performed a genome-wide screen of single nucleotide polymorphisms (SNPs) and studied its immunological consequence. METHODS Chronic hepatitis B patients (40 HBeAg-positive and 44 HBeAg-negative) treated with peginterferon alfa-2a and adefovir were genotyped for 999,091 SNPs, which were associated with HBsAg loss at week 96 (n = 9). Plasma carnitine levels were measured by tandem-mass spectrometry, and the effect of carnitine on the proliferative capacity of hepatitis B virus (HBV)-specific and non-specific CD8 T cells was studied in vitro. RESULTS One polymorphism, rs12356193 located in the SLC16A9 gene, was genome-wide significantly associated with HBsAg loss at week 96 (p = 1.84 × 10(-8)). The previously reported association of rs12356193 with lower carnitine levels was confirmed in our cohort, and baseline carnitine levels were lower in patients with HBsAg loss compared to patients with HBsAg persistence (p = 0.02). Furthermore, we demonstrated that carnitine suppressed HBV-specific CD8 T cell proliferation. CONCLUSIONS In chronic hepatitis B patients treated with peginterferon and adefovir, we identified strong associations of SLC16A9 gene variation and carnitine levels with HBsAg loss. Our results further suggest that a lower baseline plasma carnitine level increases the proliferative capacity of CD8 T cells, making patients more susceptible to the immunological effect of this treatment. These novel findings may provide new insight into factors involved in treatment-induced HBsAg loss, and play a role in the prediction of treatment outcome.
Collapse
Affiliation(s)
- Louis Jansen
- Department of Gastroenterology and Hepatology, Academic Medical Center (AMC), University of Amsterdam (UvA), Amsterdam, The Netherlands; Department of Experimental Immunology, AMC, UvA, The Netherlands
| | - Annikki de Niet
- Department of Gastroenterology and Hepatology, Academic Medical Center (AMC), University of Amsterdam (UvA), Amsterdam, The Netherlands; Department of Experimental Immunology, AMC, UvA, The Netherlands
| | - Femke Stelma
- Department of Gastroenterology and Hepatology, Academic Medical Center (AMC), University of Amsterdam (UvA), Amsterdam, The Netherlands; Department of Experimental Immunology, AMC, UvA, The Netherlands
| | - Erik P A van Iperen
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, AMC, UvA, The Netherlands; Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
| | - Karel A van Dort
- Department of Experimental Immunology, AMC, UvA, The Netherlands
| | - Marjan J Tempelmans Plat-Sinnige
- Department of Gastroenterology and Hepatology, Academic Medical Center (AMC), University of Amsterdam (UvA), Amsterdam, The Netherlands; Department of Experimental Immunology, AMC, UvA, The Netherlands
| | - R Bart Takkenberg
- Department of Gastroenterology and Hepatology, Academic Medical Center (AMC), University of Amsterdam (UvA), Amsterdam, The Netherlands
| | | | - A H Koos Zwinderman
- Department of Clinical Epidemiology, Biostatistics, and Bioinformatics, AMC, UvA, The Netherlands
| | - Uri Lopatin
- Assembly Pharmaceuticals, Bloomington, IN, USA
| | | | - Hendrik W Reesink
- Department of Gastroenterology and Hepatology, Academic Medical Center (AMC), University of Amsterdam (UvA), Amsterdam, The Netherlands.
| |
Collapse
|
298
|
Chino Y, Samukawa Y, Sakai S, Nakai Y, Yamaguchi JI, Nakanishi T, Tamai I. SGLT2 inhibitor lowers serum uric acid through alteration of uric acid transport activity in renal tubule by increased glycosuria. Biopharm Drug Dispos 2014; 35:391-404. [PMID: 25044127 PMCID: PMC4223977 DOI: 10.1002/bdd.1909] [Citation(s) in RCA: 280] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 06/24/2014] [Accepted: 07/01/2014] [Indexed: 12/13/2022]
Abstract
Sodium glucose cotransporter 2 (SGLT2) inhibitors have been reported to lower the serum uric acid (SUA) level. To elucidate the mechanism responsible for this reduction, SUA and the urinary excretion rate of uric acid (UE(UA)) were analysed after the oral administration of luseogliflozin, a SGLT2 inhibitor, to healthy subjects. After dosing, SUA decreased, and a negative correlation was observed between the SUA level and the UE(UA), suggesting that SUA decreased as a result of the increase in the UE(UA). The increase in UE(UA) was correlated with an increase in urinary D-glucose excretion, but not with the plasma luseogliflozin concentration. Additionally, in vitro transport experiments showed that luseogliflozin had no direct effect on the transporters involved in renal UA reabsorption. To explain that the increase in UE(UA) is likely due to glycosuria, the study focused on the facilitative glucose transporter 9 isoform 2 (GLUT9ΔN, SLC2A9b), which is expressed at the apical membrane of the kidney tubular cells and transports both UA and D-glucose. It was observed that the efflux of [(14) C]UA in Xenopus oocytes expressing the GLUT9 isoform 2 was trans-stimulated by 10 mm D-glucose, a high concentration of glucose that existed under SGLT2 inhibition. On the other hand, the uptake of [(14) C]UA by oocytes was cis-inhibited by 100 mm D-glucose, a concentration assumed to exist in collecting ducts. In conclusion, it was demonstrated that the UE(UA) could potentially be increased by luseogliflozin-induced glycosuria, with alterations of UA transport activity because of urinary glucose.
Collapse
Affiliation(s)
- Yukihiro Chino
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKakuma-machi, Kanazawa, 920-1192, Japan
- Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd1-403 Yoshino-cho, Kita-ku, Saitama, 331-9530, Japan
| | - Yoshishige Samukawa
- Development Management, Taisho Pharmaceutical Co., Ltd3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Soichi Sakai
- Clinical Research, Taisho Pharmaceutical Co., Ltd3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Yasuhiro Nakai
- Development Management, Taisho Pharmaceutical Co., Ltd3-24-1, Takada, Toshima-ku, Tokyo, 170-8633, Japan
| | - Jun-ichi Yamaguchi
- Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd1-403 Yoshino-cho, Kita-ku, Saitama, 331-9530, Japan
| | - Takeo Nakanishi
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKakuma-machi, Kanazawa, 920-1192, Japan
| | - Ikumi Tamai
- Department of Membrane Transport and Biopharmaceutics, Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKakuma-machi, Kanazawa, 920-1192, Japan
| |
Collapse
|
299
|
Prestin K, Wolf S, Feldtmann R, Hussner J, Geissler I, Rimmbach C, Kroemer HK, Zimmermann U, Meyer zu Schwabedissen HE. Transcriptional regulation of urate transportosome member SLC2A9 by nuclear receptor HNF4α. Am J Physiol Renal Physiol 2014; 307:F1041-51. [PMID: 25209865 DOI: 10.1152/ajprenal.00640.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal tubular handling of urate is realized by a network of uptake and efflux transporters, including members of drug transporter families such as solute carrier proteins and ATP-binding cassette transporters. Solute carrier family 2, member 9 (SLC2A9), is one key factor of this so called "urate transportosome." The aim of the present study was to understand the transcriptional regulation of SLC2A9 and to test whether identified factors might contribute to a coordinated transcriptional regulation of the transporters involved in urate handling. In silico analysis and cell-based reporter gene assays identified a hepatocyte nuclear factor (HNF)4α-binding site in the promoter of SLC2A9 isoform 1, whose activity was enhanced by transient HNF4α overexpression, whereas mutation of the binding site diminished activation. HNF4α overexpression induced endogenous SLC2A9 expression in vitro. The in vivo role of HNF4α in the modulation of renal SLC2A9 gene expression was supported by findings of quantitative real-time RT-PCR analyses and chromatin immunoprecipitation assays. Indeed, mRNA expression of SLC2A9 and HNF4α in human kidney samples was significantly correlated. We also showed that in renal clear cell carcinoma, downregulation of HNF4α mRNA and protein expression was associated with a significant decline in expression of the transporter. Taken together, our data suggest that nuclear receptor family member HNF4α contributes to the transcriptional regulation of SLC2A9 isoform 1. Since HNF4α has previously been assumed to be a modulator of several urate transporters, our findings support the notion that there could be a transcriptional network providing synchronized regulation of the functional network of the urate transportosome.
Collapse
Affiliation(s)
- Katharina Prestin
- University of Basel, Department of Pharmaceutical Sciences, Biopharmacy, Basel, Switzerland
| | - Stephanie Wolf
- University Medicine, Ernst Moritz Arndt University Greifswald, Center of Drug Absorption and Transport, Institute of Pharmacology, Greifswald, Germany
| | - Rico Feldtmann
- University Medicine, Ernst Moritz Arndt University Greifswald, Center of Drug Absorption and Transport, Institute of Pharmacology, Greifswald, Germany
| | - Janine Hussner
- University of Basel, Department of Pharmaceutical Sciences, Biopharmacy, Basel, Switzerland
| | - Ingrid Geissler
- University Medicine, Ernst Moritz Arndt University Greifswald, Center of Drug Absorption and Transport, Institute of Pharmacology, Greifswald, Germany
| | - Christian Rimmbach
- University Medicine, Ernst Moritz Arndt University Greifswald, Center of Drug Absorption and Transport, Institute of Pharmacology, Greifswald, Germany
| | - Heyo K Kroemer
- University of Goettingen, Medical Faculty, Goettingen, Germany; and
| | - Uwe Zimmermann
- University Medicine, Ernst Moritz Arndt University Greifswald, Department of Urology, Greifswald, Germany
| | | |
Collapse
|
300
|
Abstract
Over the last decade, the biologic interference of uric acid with the cardiovascular (CV) system and the kidney has been intensively investigated, and several experimental studies in animal models and in vitro documented that hyperuricemia may trigger hypertension and incite endothelial dysfunction, vascular damage and renal disease. A substantial proportion of epidemiological studies are compatible with the hypothesis that hyperuricemia may be noxious to the CV system and the kidney as well. However, there are still no well-powered trials testing whether uric acid-lowering interventions may reduce BP or attenuate the risk for adverse CV and renal outcomes. Evidence still remains largely insufficient to recommend changes in the current policy of not prescribing uric acid-lowering drugs to individuals with asymptomatic hyperuricemia.
Collapse
|