101
|
Khaliq OP, Konoshita T, Moodely J, Ramsuran V, Naicker T. Gene polymorphisms of uric acid are associated with pre-eclampsia in South Africans of African ancestry. Hypertens Pregnancy 2020; 39:103-116. [PMID: 32255363 DOI: 10.1080/10641955.2020.1741608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objectives: To investigate the association of uric acid gene polymorphisms and Pre-eclampsia.Methods: 637 women of African ancestry [280 controls, 357 pre-eclampsia (early-onset = 187, late-onset = 170]) retrospectively. The rs505802, rs1212986, and rs1014290 SNPs were genotyped from purified DNA using real-time PCR.Results: CT genotype (rs505802) was higher in pre-eclampsia [Adjusted p = 0.028*: OR (95% CI) = 1.73 (1.258-2.442)] and late-onset pre-eclampsia [Adjusted p = 0.027*: OR (95% CI) = 1.75 (1.165-2.2628)] than controls. CT genotype (rs1014290) was higher in early-onset pre-eclampsia [Adjusted p-value = 0.040*: OR (95% CI) = 1.60 (1.102-2.325)] than controls.Conclusion: The genotyped rs505802 and rs1014290 are significantly associated with pre-eclampsia.
Collapse
Affiliation(s)
- Olive P Khaliq
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Tadashi Konoshita
- Third Department of Internal Medicine, University of Fukui Faculty of Medicine Sciences, Fukui, Japan
| | - Jagidesa Moodely
- Department of Obstetrics and Gynecology and Women's Health and HIV Research Group, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Veron Ramsuran
- KwaZulu-Natal Research Innovation and Sequencing Platform, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
102
|
Rafiullah M, Siddiqui K, Al-Rubeaan K. Association between serum uric acid levels and metabolic markers in patients with type 2 diabetes from a community with high diabetes prevalence. Int J Clin Pract 2020; 74:e13466. [PMID: 31854061 DOI: 10.1111/ijcp.13466] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The importance of uric acid has been increasingly appreciated because of its association with the development of diabetes mellitus and related diseases. OBJECTIVE This study was undertaken to evaluate the association of serum uric acid (SUA) level with different clinical and biochemical parameters in patients with type 2 diabetes from Saudi Arabia. METHODS Clinical and biochemical data from the patients were obtained and assessed in a cross-sectional design. Relationships between SUA level and various clinical and biochemical parameters were analysed. RESULTS SUA level was positively associated with increased incidence of cardiovascular diseases (CVD) in patients with abnormal eGFR (<90 mL/min/1.73 m2 ). HbA1c was found to be inversely associated with hyperuricemia in patients with normal eGFR level (≥90 mL/min/1.73 m2 ). Incidence of metabolic syndrome did not show any relationship with SUA level. However, the incidence of hypertension, a component of metabolic syndrome, was significantly higher among patients with hyperuricemia. Waist circumference and serum triglycerides were higher, whereas serum high-density lipoprotein level was lower in patients with higher SUA level. Patients with hyperuricemia had higher incidence of CVDs than those of the normouricemic group. CONCLUSION SUA level was positively associated with incidence of cardiovascular diseases CVD in patients with abnormal eGFR. HbA1c correlated significantly with SUA level in patients with normal eGFR. Incidence of metabolic syndrome did not show any association with SUA level. Incidence of hypertension, waist circumference and serum triglycerides were significantly higher and serum high-density lipoprotein level was lower in patients with higher SUA level.
Collapse
Affiliation(s)
- Mohamed Rafiullah
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Khalid Siddiqui
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Khalid Al-Rubeaan
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
103
|
Gender-related differences in serum uric acid in treated hypertensive patients from central and east European countries: findings from the Blood Pressure control rate and CArdiovascular Risk profilE study. J Hypertens 2020; 37:380-388. [PMID: 30074564 DOI: 10.1097/hjh.0000000000001908] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Hyperuricemia has been associated with high blood pressure (BP) values, diabetes mellitus, metabolic syndrome and chronic kidney disease (CKD). In the present study, we assessed the gender-related relationships between serum uric acid (SUA) and cardionephrometabolic variables in central and east European hypertensive patients. METHODS A total of 3206 treated hypertensive patients with available SUA levels from the BP-CARE study was analyzed. Correlations among SUA, BP values, BP control, diabetes mellitus, metabolic syndrome and CKD were performed according to gender. RESULTS Twenty-five percent of the whole population showed hyperuricemia (28% in women and 23% in men). These patients were older and showed a greater burden of cardiovascular risk factors (high BP, BMI, glucose, total cholesterol and triglyceridemia). They also showed a greater prevalence of metabolic syndrome, diabetes mellitus, rate of uncontrolled BP, more than high cardiovascular risk and CKD. Prevalence of metabolic syndrome and uncontrolled BP was similar in normouricemic and hyperuricemic women, the latter displaying a higher prevalence of diabetes mellitus, high cardiovascular risk and CKD. Hyperuricemic men were characterized by a greater prevalence of metabolic syndrome, diabetes mellitus, high cardiovascular risk, rate of uncontrolled BP and CKD but not polytherapy. Logistic regression analysis showed that none of the evaluated variables, except CKD, displayed SUA as significant covariate. CONCLUSION Our findings provide evidence that a high prevalence of hyperuricemia occurs in hypertensive patients from central and east Europe. The data also show that gender-related differences in the association between SUA and cardionephrometabolic variables exist. This is also the case for the relationships between SUA and CKD.
Collapse
|
104
|
Nakashima A, Ichida K, Ohkido I, Yokoyama K, Matsuo H, Ohashi Y, Takada T, Nakayama A, Suzuki H, Shinomiya N, Urashima M, Yokoo T. Dysfunctional ABCG2 gene polymorphisms are associated with serum uric acid levels and all-cause mortality in hemodialysis patients. Hum Cell 2020; 33:559-568. [PMID: 32180207 PMCID: PMC7324430 DOI: 10.1007/s13577-020-00342-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Dysfunctional variants of ATP-binding cassette transporter subfamily G member 2 (ABCG2), a urate transporter in the kidney and intestine, are the major causes of hyperuricemia and gout. A recent study found that ABCG2 is a major transporter of uremic toxins; however, few studies have investigated the relationship between ABCG2 gene polymorphisms and mortality. This prospective cohort study of 1214 hemodialysis patients investigated the association between serum uric acid levels and ABCG2 genotype and mortality. Genotyping of dysfunctional ABCG2 variants, Q126X (rs72552713) and Q141K (rs2231142), was performed using the patients’ DNA. During the study period, 220 patients died. Lower serum uric acid levels were associated with higher mortality (hazard ratio [HR] 1.89, 95% confidence interval [CI] 1.14–3.10, P ≤ 0.001). ABCG2 dysfunction, estimated by genetic variants, had a significant positive association with serum uric acid levels (full function: 7.4 ± 1.2 mg/dl, 3/4 function: 7.9 ± 1.3 mg/dl, 1/2 function: 8.2 ± 1.4 mg/dl, ≤ 1/4 function: 8.7 ± 1.3 mg/dl, P ≤ 0.001). This association remained significant on multiple regression analysis. The Cox proportional hazard analysis indicated that the ABCG2 ≤ 1/4 function type was significantly associated with higher mortality (HR 6.66, 95% CI 2.49 to 17.8, P ≤ 0.001) than the other function types. These results showed that ABCG2 plays a physiologically important role in uric acid excretion, and that ABCG2 dysfunction is a risk factor for mortality in hemodialysis patients.
Collapse
Affiliation(s)
- Akio Nakashima
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Kimiyoshi Ichida
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan. .,Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| | - Ichiro Ohkido
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Keitaro Yokoyama
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yuki Ohashi
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Mitsuyoshi Urashima
- Division of Molecular Epidemiology, Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
105
|
Maloberti A, Giannattasio C, Bombelli M, Desideri G, Cicero AFG, Muiesan ML, Rosei EA, Salvetti M, Ungar A, Rivasi G, Pontremoli R, Viazzi F, Facchetti R, Ferri C, Bernardino B, Galletti F, D'Elia L, Palatini P, Casiglia E, Tikhonoff V, Barbagallo CM, Verdecchia P, Masi S, Mallamaci F, Cirillo M, Rattazzi M, Pauletto P, Cirillo P, Gesualdo L, Mazza A, Volpe M, Tocci G, Iaccarino G, Nazzaro P, Lippa L, Parati G, Dell'Oro R, Quarti-Trevano F, Grassi G, Virdis A, Borghi C. Hyperuricemia and Risk of Cardiovascular Outcomes: The Experience of the URRAH (Uric Acid Right for Heart Health) Project. High Blood Press Cardiovasc Prev 2020; 27:121-128. [PMID: 32157643 DOI: 10.1007/s40292-020-00368-z] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/04/2020] [Indexed: 12/17/2022] Open
Abstract
The latest European Guidelines of Arterial Hypertension have officially introduced uric acid evaluation among the cardiovascular risk factors that should be evaluated in order to stratify patient's risk. In fact, it has been extensively evaluated and demonstrated to be an independent predictor not only of all-cause and cardiovascular mortality, but also of myocardial infraction, stroke and heart failure. Despite the large number of studies on this topic, an important open question that still need to be answered is the identification of a cardiovascular uric acid cut-off value. The actual hyperuricemia cut-off (> 6 mg/dL in women and 7 mg/dL in men) is principally based on the saturation point of uric acid but previous evidence suggests that the negative impact of cardiovascular system could occur also at lower levels. In this context, the Working Group on uric acid and CV risk of the Italian Society of Hypertension has designed the Uric acid Right for heArt Health project. The primary objective of this project is to define the level of uricemia above which the independent risk of CV disease may increase in a significantly manner. In this review we will summarize the first results obtained and describe the further planned analysis.
Collapse
Affiliation(s)
- Alessandro Maloberti
- Cardiology IV, "A.De Gasperis" Department, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy. .,Health Science Department, Milano-Bicocca University, Milan, Italy.
| | - C Giannattasio
- Cardiology IV, "A.De Gasperis" Department, Ospedale Niguarda Ca' Granda, Piazza Ospedale Maggiore 3, 20159, Milan, Italy.,Health Science Department, Milano-Bicocca University, Milan, Italy
| | - M Bombelli
- Health Science Department, Milano-Bicocca University, Milan, Italy.,Clinica Medica, San Gerardo Hospital, Monza, Italy
| | - G Desideri
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - A F G Cicero
- Department of Medical and Surgical Science, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - M L Muiesan
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - E A Rosei
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - M Salvetti
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - A Ungar
- Department of Geriatric and Intensive Care Medicine, Careggi Hospital and University of Florence, Florence, Italy
| | - G Rivasi
- Department of Geriatric and Intensive Care Medicine, Careggi Hospital and University of Florence, Florence, Italy
| | - R Pontremoli
- Department of Internal Medicine, University of Genoa and Policlinico SanMartino, Genoa, Italy
| | - F Viazzi
- Department of Internal Medicine, University of Genoa and Policlinico SanMartino, Genoa, Italy
| | - R Facchetti
- Health Science Department, Milano-Bicocca University, Milan, Italy
| | - C Ferri
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - B Bernardino
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - F Galletti
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Naples, Italy
| | - L D'Elia
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples Medical School, Naples, Italy
| | - P Palatini
- Studium Patavinum, Department of Medicine, University of Padua, Padua, Italy
| | - E Casiglia
- Studium Patavinum, Department of Medicine, University of Padua, Padua, Italy
| | - V Tikhonoff
- Department of Medicine, University of Padua, Padua, Italy
| | - C M Barbagallo
- Biomedical Department of Internal Medicine and Specialistics, University of Palermo, Palermo, Italy
| | - P Verdecchia
- Hospital S. Maria della Misericordia, Perugia, Italy
| | - S Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - F Mallamaci
- Reggio Cal Unit, CNR-IFC, Clinical Epidemiology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | - M Cirillo
- Department of Public Health, "Federico II" University of Naples, Naples, Italy
| | - M Rattazzi
- Department of Medicine, University of Padua, Padua, Italy.,Medicina Interna I, Ca' Foncello University Hospital, Treviso, Italy
| | - P Pauletto
- Medicina Interna I, Ca' Foncello University Hospital, Treviso, Italy
| | - P Cirillo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, "Aldo Moro" University of Bari, Bari, Italy
| | - L Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, "Aldo Moro" University of Bari, Bari, Italy
| | - A Mazza
- Department of Internal Medicine, Santa Maria della Misericordia General Hospital, AULSS 5 Polesana, Rovigo, Italy
| | - M Volpe
- Division of Cardiology, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University of Rome Sapienza, Sant'Andrea Hospital, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - G Tocci
- Division of Cardiology, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University of Rome Sapienza, Sant'Andrea Hospital, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - G Iaccarino
- Department of Advanced Biomedical Sciences, "Federico II" University of Naples, Naples, Italy
| | - P Nazzaro
- Department of Medical Basic Sciences, Neurosciences and Sense Organs, University of Bari Medical School, Bari, Italy
| | - L Lippa
- Italian Society of General Medicine (SIMG), Avezzano, L'Aquila, Italy
| | - G Parati
- Health Science Department, Milano-Bicocca University, Milan, Italy.,Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - R Dell'Oro
- Health Science Department, Milano-Bicocca University, Milan, Italy.,Clinica Medica, San Gerardo Hospital, Monza, Italy
| | - F Quarti-Trevano
- Health Science Department, Milano-Bicocca University, Milan, Italy.,Clinica Medica, San Gerardo Hospital, Monza, Italy
| | - G Grassi
- Health Science Department, Milano-Bicocca University, Milan, Italy.,Clinica Medica, San Gerardo Hospital, Monza, Italy
| | - A Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - C Borghi
- Department of Medical and Surgical Science, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | | |
Collapse
|
106
|
Rebora P, Andreano A, Triglione N, Piccinelli E, Palazzini M, Occhi L, Grassi G, Valsecchi MG, Giannattasio C, Maloberti A. Association between uric acid and pulse wave velocity in hypertensive patients and in the general population: a systematic review and meta-analysis. Blood Press 2020; 29:220-231. [PMID: 32138547 DOI: 10.1080/08037051.2020.1735929] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Purpose: The association between serum uric acid (SUA) and pulse wave velocity (PWV), has been extensively evaluated but with some discrepancies in results. A further limitation refers to the fact that only few data were analyzed taking into account the possible effects of gender. The purpose of this study was to estimate the association between SUA and arterial stiffness in general population and hypertensive patients, as a whole population and as divided by gender, by pooling results from existing studies.Materials and methods: Carotid-femoral and brachial-ankle PWV (cf- and ba-PWV) have been analyzed separately and subgroup analyses by gender are reported. Among 692 potentially relevant works, 24 articles were analyzed.Results: Seven studies referred to cf-PWV in the general population with an overall positive association at adjusted analysis for both males and females (beta regression coefficient (ß): 0.07; 95%CI: 0.03; 0.11 and ß: 0.06; 95%CI: 0.03; 0.09, respectively). Twelve studies referred to ba-PWV in the general population with the finding of a positive association at adjusted analysis for females (ß: 0.04; 95% confidence interval (CI): 0.01;0.07), but not for males (ß: 0.13; 95%CI: -0.09; 0.34). In hypertensive patients only four studies evaluated cf-PWV and one ba-PWV with only one study (with cf-PWV) finding positive association.Conclusion: The association between SUA and cf-PWV resulted significant in general population in both males and females while it was only significant for female regarding ba-PWV. Furthermore, the few available studies found no significant relationship between SUA and both cf- and ba-PWV in hypertensive subjects.
Collapse
Affiliation(s)
- Paola Rebora
- Center of Biostatistics for Clinical Epidemiology, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Anita Andreano
- Center of Biostatistics for Clinical Epidemiology, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Nicola Triglione
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Enrico Piccinelli
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Matteo Palazzini
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Lucia Occhi
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Guido Grassi
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Maria Grazia Valsecchi
- Center of Biostatistics for Clinical Epidemiology, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Cristina Giannattasio
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy.,Cardiology IV, "A.De Gasperis" Department, ASTT GOM Niguarda Ca' Granda, Milan, Italy
| | - Alessandro Maloberti
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy.,Cardiology IV, "A.De Gasperis" Department, ASTT GOM Niguarda Ca' Granda, Milan, Italy
| |
Collapse
|
107
|
Xia X, Jin J, Chen ZJ, Zhou Z, Chen H, Zhang C, Zhang L, Sun L. Unraveling the genetic causes in large pedigrees with gout by whole‑exome sequencing. Int J Mol Med 2020; 45:1047-1058. [PMID: 32124961 PMCID: PMC7053847 DOI: 10.3892/ijmm.2020.4501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/08/2020] [Indexed: 11/06/2022] Open
Abstract
Gout is a common type of inflammatory arthritis that is clinically and genetically heterogeneous. The genetic aetiology remains unclear, and mainly relies on previous genome-wide association studies focused on sporadic cases. The present study aimed to identify the genetic basis of gout in three families using whole-exome sequencing (WES). WES was performed in the probands, and family members were involved in the co-segregation analysis. In total, three deleterious rare or novel missense mutations were identified in ATP-binding cassette super-family G member 2 (ABCG2), protein kinase CGMP-dependent 2 (PRKG2) and adrenoceptor β3 (ADRB3) genes in three different families. In addition, certain gout-associated candidate genes were revealed to be shared among the co-expression and protein-protein interaction (PPI) networks of ABCG2, PRKG2 and ADRB3. Furthermore, the disease ontology analysis of the genes present in the co-expression network exhibited significant (P<0.05) enrichment in hyperuricemia, gout, cardiovascular system disease and metabolic disease. In addition, genes involved in the PPI network were significantly enriched in the purine nucleoside monophosphate biosynthetic process, urate transport and biological processes associated with glycose metabolism. Collectively, to the best of our knowledge, the present study was the first to use WES to identify three candidate rare or novel deleterious mutations in three families with gout. The present results provided novel insights that may improve the current understanding of the molecular genetic basis underlying gout. Importantly, the present results may facilitate the improvement of clinical diagnosis and the development of novel personalized therapies.
Collapse
Affiliation(s)
- Xiaoru Xia
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325003, P.R. China
| | - Jing Jin
- Zhejiang Center for Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Zhen-Ji Chen
- Division of Ophthalmic Genetics, Laboratory for Stem Cell and Retinal Regeneration, Institute of Stem Cell Research, The Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhenni Zhou
- Department of Internal Medicine, Yueqing People's Hospital, Yueqing, Wenzhou, Zhejiang 325600, P.R. China
| | - Hui Chen
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Chunwu Zhang
- Department of Injury Orthopaedics, The First Affiliated Hospital of Wenzhou University, Wenzhou, Zhejiang 325023, P.R. China
| | - Linhua Zhang
- Department of Clinical Laboratory, Yuhuan People's Hospital, Taizhou, Zhejiang 317600, P.R. China
| | - Li Sun
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325003, P.R. China
| |
Collapse
|
108
|
Abstract
BACKGROUND Formate is a one-carbon molecule at the crossroad between cellular and whole body metabolism, between host and microbiome metabolism, and between nutrition and toxicology. This centrality confers formate with a key role in human physiology and disease that is currently unappreciated. SCOPE OF REVIEW Here we review the scientific literature on formate metabolism, highlighting cellular pathways, whole body metabolism, and interactions with the diet and the gut microbiome. We will discuss the relevance of formate metabolism in the context of embryonic development, cancer, obesity, immunometabolism, and neurodegeneration. MAJOR CONCLUSIONS We will conclude with an outlook of some open questions bringing formate metabolism into the spotlight.
Collapse
Affiliation(s)
| | - Johannes Meiser
- Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Alexei Vazquez
- Cancer Research UK Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
109
|
Molecular characterization of the orphan transporter SLC16A9, an extracellular pH- and Na+-sensitive creatine transporter. Biochem Biophys Res Commun 2020; 522:539-544. [DOI: 10.1016/j.bbrc.2019.11.137] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023]
|
110
|
Keerman M, Yang F, Hu H, Wang J, Wang F, Li Z, Yuan J, Yao P, Zhang X, Guo H, Yang H, He M. Mendelian randomization study of serum uric acid levels and diabetes risk: evidence from the Dongfeng-Tongji cohort. BMJ Open Diabetes Res Care 2020; 8:8/1/e000834. [PMID: 32111716 PMCID: PMC7050304 DOI: 10.1136/bmjdrc-2019-000834] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/03/2020] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Limited Mendelian randomization (MR) studies have assessed the causal relationship between serum uric acid levels and diabetes risk. Here we investigated causality between the serum uric acid concentration and diabetes risk in Chinese population. RESEARCH DESIGN AND METHODS The observational analysis, based on the Dongfeng-Tongji prospective cohort (n=15 195) we tested the association of serum uric acid levels with incident diabetes risk. In the instrumental variable analysis, we examined the association of the genetic risk score (GRS) of serum uric acid with diabetes risk in case-control design (2539 cases and 4595 controls) via MR analysis. RESULTS During a mean (SD) follow-up of 4.5 (0.5) years, 1156 incident diabetes cases were identified. Compared with those in the lowest quintile of serum uric acid levels, the HRs of incident diabetes were 1.19 (95% CI 0.96 to 1.48), 1.12 (95% CI 0.90 to 1.40), 1.38 (95% CI 1.12 to 1.70), and 1.51 (95% CI 1.23 to 1.87) for Q2, Q3, Q4 and Q5, respectively (P-trend <0.001). The GRS was strongly associated with serum uric acid levels (β=0.17, 95% CI 0.15 to 0.19; P=2.81×10-67). However, no significant association was observed between the GRS and diabetes risk (OR=1.01, 95 CI 0.95 to 1.06; P=0.75). CONCLUSIONS Even though serum uric acid levels were significantly associated with increased incident diabetes risk, the results did not provide evidence for a causal relationship between them.
Collapse
Affiliation(s)
- Mulatibieke Keerman
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Yang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Hu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Wang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoyang Li
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yuan
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Guo
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Handong Yang
- Department of Cardiovascular Disease, Dongfeng Motor Corporation General Hospital, Shiyan, China
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental and Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
111
|
Whole-Exome Sequencing Reveals a Rare Missense Variant in SLC16A9 in a Pedigree with Early-Onset Gout. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4321419. [PMID: 32090094 PMCID: PMC7013288 DOI: 10.1155/2020/4321419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 12/28/2019] [Accepted: 01/10/2020] [Indexed: 02/06/2023]
Abstract
Gout is a common inflammatory arthritis triggered by monosodium urate deposition after longstanding hyperuricemia. In the general community, the disease is largely polygenic in genetic architecture, with many polymorphisms having been identified in gout or urate-associated traits. In a small proportion of cases, rare high penetrant mutations associated with monogenic segregation of the disease in families have been demonstrated to be disease causative. In this study, we recruited a two-generation pedigree with early-onset gout. To elucidate the genetic predisposition, whole-exome sequencing (WES) was performed. After comprehensive variant analyses and cosegregation testing, we identified a missense variant (c.277C>A, p.L93M) in SLC16A9, an extremely rare variant in genetic databases. Moreover, in silico assessments showed strong pathogenicity. This variant cosegregated with the disease phenotype perfectly in the family and is located in a highly conserved functional domain. A few studies supported our results of the association between SLC16A9 and gout and serum urate levels. In conclusion, we provide the first evidence for the association of rare missense in SLC16A9 with early-onset gout. These findings not only expand our current understanding of gout but also may have further implications for the treatment and prevention of gout.
Collapse
|
112
|
Dong Z, Zhou J, Jiang S, Li Y, Zhao D, Yang C, Ma Y, He H, Ji H, Jin L, Zou H, Wang J. Epistatic interaction between PKD2 and ABCG2 influences the pathogenesis of hyperuricemia and gout. Hereditas 2020; 157:2. [PMID: 32000861 PMCID: PMC6986014 DOI: 10.1186/s41065-020-0116-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/14/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Genetic background affects serum urate concentration and gout risk, especially regarding these variants in the urate-transporter gene ABCG2. However, the role of epistasis between PKD2 and ABCG2 on the pathogenesis of gout is poorly understood. Here we assess this epistatic interaction in the progression from elevated serum urate to gout. RESULTS We identified two epistatic interaction pairs (rs2728121: rs1481012 and rs2728121: rs2231137) were associated with urate levels in 4914 Chinese individuals (Pint = 0.018 and 0.004, respectively). Using subgroup analysis for gender and BMI, we found the degree of associations was varied by gender and BMI. The SNP pair rs2728121:rs1481012 influenced urate levels in females and overweight subjects (Pint = 0.006 and 0.022, respectively), but rs2728121:rs2231137 did in males, overweight and normal-weight subjects (Pint = 0.017, 0.047 and 0.013, respectively). Consistent results were also observed in associations between these epistatic interactions with hyperuricemia. Next, the SNP pair rs2728121:rs2231137 was identified to influence the development of gout from both hyperuricemia and healthy (Pint = 0.035 and 0.001, respectively), especially in males (Pint = 0.030 and 0.001, respectively). Furthermore, we demonstrated that interacting regions were enriched by regulatory elements. Finally, we observed a strong gene co-expression pattern between PKD2 and ABCG2 (r = 0.743, P = 5.83E-06). CONCLUSION Our findings indicate epistasis between PKD2 and ABCG2 influence serum urate concentrations, hyperuricemia and gout risk, thus providing insight into the pathogenesis of gout.
Collapse
Affiliation(s)
- Zheng Dong
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai, 200438, People's Republic of China
| | - Jingru Zhou
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai, 200438, People's Republic of China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai, 200438, People's Republic of China
| | - Yuan Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai, 200438, People's Republic of China
| | - Dongbao Zhao
- Division of Rheumatology and Immunology, Changhai Hospital, Shanghai, China
| | - Chengde Yang
- Division of Rheumatology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai, 200438, People's Republic of China
| | - Hongjun He
- Division of Rheumatology, Taixing People's Hospital, Jiangsu Province, China
| | - Hengdong Ji
- Division of Rheumatology, Taizhou People's Hospital, Jiangsu Province, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai, 200438, People's Republic of China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu Province, China
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, People's Republic of China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University Jiangwan Campus, 2005 Songhu Road, Shanghai, 200438, People's Republic of China.
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu Province, China.
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China.
| |
Collapse
|
113
|
Schumann T, König J, Henke C, Willmes DM, Bornstein SR, Jordan J, Fromm MF, Birkenfeld AL. Solute Carrier Transporters as Potential Targets for the Treatment of Metabolic Disease. Pharmacol Rev 2020; 72:343-379. [PMID: 31882442 DOI: 10.1124/pr.118.015735] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The solute carrier (SLC) superfamily comprises more than 400 transport proteins mediating the influx and efflux of substances such as ions, nucleotides, and sugars across biological membranes. Over 80 SLC transporters have been linked to human diseases, including obesity and type 2 diabetes (T2D). This observation highlights the importance of SLCs for human (patho)physiology. Yet, only a small number of SLC proteins are validated drug targets. The most recent drug class approved for the treatment of T2D targets sodium-glucose cotransporter 2, product of the SLC5A2 gene. There is great interest in identifying other SLC transporters as potential targets for the treatment of metabolic diseases. Finding better treatments will prove essential in future years, given the enormous personal and socioeconomic burden posed by more than 500 million patients with T2D by 2040 worldwide. In this review, we summarize the evidence for SLC transporters as target structures in metabolic disease. To this end, we identified SLC13A5/sodium-coupled citrate transporter, and recent proof-of-concept studies confirm its therapeutic potential in T2D and nonalcoholic fatty liver disease. Further SLC transporters were linked in multiple genome-wide association studies to T2D or related metabolic disorders. In addition to presenting better-characterized potential therapeutic targets, we discuss the likely unnoticed link between other SLC transporters and metabolic disease. Recognition of their potential may promote research on these proteins for future medical management of human metabolic diseases such as obesity, fatty liver disease, and T2D. SIGNIFICANCE STATEMENT: Given the fact that the prevalence of human metabolic diseases such as obesity and type 2 diabetes has dramatically risen, pharmacological intervention will be a key future approach to managing their burden and reducing mortality. In this review, we present the evidence for solute carrier (SLC) genes associated with human metabolic diseases and discuss the potential of SLC transporters as therapeutic target structures.
Collapse
Affiliation(s)
- Tina Schumann
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Jörg König
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Diana M Willmes
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Stefan R Bornstein
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Jens Jordan
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Martin F Fromm
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| |
Collapse
|
114
|
Lee YH, Song GG. The Uric Acid and Gout have No Direct Causality With Osteoarthritis: A Mendelian Randomization Study. JOURNAL OF RHEUMATIC DISEASES 2020. [DOI: 10.4078/jrd.2020.27.2.88] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
115
|
Stewart DJ, Langlois V, Noone D. Hyperuricemia and Hypertension: Links and Risks. Integr Blood Press Control 2019; 12:43-62. [PMID: 31920373 PMCID: PMC6935283 DOI: 10.2147/ibpc.s184685] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022] Open
Abstract
Hyperuricemia has long been recognized to be associated with increased cardiovascular risk, including risk of developing hypertension. Epidemiological findings suggest that the link with hypertension is stronger in children and adolescents. Uric acid acts as a strong antioxidant compound in the extracellular environment but has pro-inflammatory effects within the intracellular setting. A chronic phase of microvascular injury is known to occur after prolonged periods of hyperuricemia. This is proposed to contribute to afferent arteriolopathy and elevation of blood pressure that may become unresponsive to uric acid-lowering therapies over time. Studies have struggled to infer direct causality of hyperuricemia due to a vast number of confounders including body mass index. The aim of this review is to present the available data and highlight the need for large scale prospective randomized controlled trials in this area. At present, there is limited evidence to support a role for uric acid-lowering therapies in helping mitigate the risk of hypertension.
Collapse
Affiliation(s)
- Douglas J Stewart
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Valerie Langlois
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada.,Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Damien Noone
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada.,Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
116
|
Narang RK, Vincent Z, Phipps-Green A, Stamp LK, Merriman TR, Dalbeth N. Population-specific factors associated with fractional excretion of uric acid. Arthritis Res Ther 2019; 21:234. [PMID: 31718705 PMCID: PMC6852918 DOI: 10.1186/s13075-019-2016-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/25/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Reduced renal clearance of uric acid is a major contributor to hyperuricemia. The aim of this study was to examine clinical and genetic variables associated with fractional excretion of uric acid (FEUA). METHODS Participants (with and without gout) in the Genetics of Gout in Aotearoa study with available genotyping and FEUA data were included (n = 1713). Ten FEUA-associated loci detected within a genome-wide association study for serum urate in a European population were analysed. A polygenic score for FEUA was calculated in each ancestry group to model the cumulative effects of the genetic variants on FEUA. Associations between FEUA and both clinical variables and polygenic score were tested using linear regression models. RESULTS The mean (SD) FEUA was 5.13 (2.70) % in Eastern Polynesian participants, 4.70 (5.89) % in Western Polynesian participants, and 5.89 (2.73) % in New Zealand European participants. Although association with FEUA was observed for SLC2A9 rs11942223 in New Zealand European participants (P = 2.39 × 10- 8), this association was not observed in Eastern or Western Polynesian participants. The polygenic score was positively associated with FEUA in all ancestry groups. In New Zealand European participants, body mass index, diuretic use, polygenic score, and male sex were associated with FEUA and explained 22% of FEUA variance in the regression model. In Eastern and Western Polynesian participants, the tested variables explained 10% and 4% of FEUA variance respectively. CONCLUSIONS Both clinical and genetic variables contribute to renal clearance of uric acid. SLC2A9 exerts effects on FEUA variance in people of European ancestry, but not in those of Polynesian ancestry. There is a large unexplained variance in FEUA, particularly in people of Polynesian ancestry.
Collapse
Affiliation(s)
- Ravi K Narang
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Zoe Vincent
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Amanda Phipps-Green
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9012, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago, 2 Riccarton Avenue, Christchurch, 8140, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9012, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| |
Collapse
|
117
|
Liu Z, Su X, Xiao M, Zhou P, Guo J, Huang Y, Zhan Y. Association between Eating Away from Home and Hyperuricemia: A Population-Based Nationwide Cross-Sectional Study in China. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2792681. [PMID: 31687384 PMCID: PMC6794973 DOI: 10.1155/2019/2792681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/20/2019] [Accepted: 09/16/2019] [Indexed: 01/29/2023]
Abstract
Hyperuricemia (HU) is a risk factor for different kinds of chronic noncommunicable diseases, and eating away from home (EAFH) may play an important role in their development, which has been ignored greatly so far. This study aimed to investigate the association between EAFH and HU in different models. A cross-sectional study involving 8,322 participants of the China Health and Nutrition Survey (CHNS) was conducted. Logistic regression models were used to analyze the data. We found that participants who consumed more away-from-home food had a higher risk for HU, and the adjusted odds ratio (aOR) and 95% confidence interval (CI) (for each increment in grades of EAFH) were 1.11 (1.02, 1.20) in a multiadjusted model (adjusted for age, gender, province, net individual income, body mass index, smoking, leisure-time physical activities, energy intake, and sleep duration). As for stratified analyses, the aOR (95% CI) of EAFH was 1.12 (1.01, 1.24) for men and 1.06 (0.92, 1.21) for women. Similar results can be found in the middle-aged and obese population, with aOR (95% CI) of EAFH as 1.17 (1.05, 1.30) and 1.15 (1.03, 1.29), respectively. In conclusion, EAFH is positively associated with the prevalence of HU.
Collapse
Affiliation(s)
- Zifeng Liu
- Department of Clinical Data Center, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiaoting Su
- Department of Health Policy and Management, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Mianli Xiao
- Department of Clinical Data Center, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Peien Zhou
- Department of Health Policy and Management, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jianwei Guo
- Department of Health Policy and Management, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yixiang Huang
- Department of Health Policy and Management, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yiqiang Zhan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm 17177, Sweden
| |
Collapse
|
118
|
Genetically determined serum urate levels and cardiovascular and other diseases in UK Biobank cohort: A phenome-wide mendelian randomization study. PLoS Med 2019; 16:e1002937. [PMID: 31626644 PMCID: PMC6799886 DOI: 10.1371/journal.pmed.1002937] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The role of urate in cardiovascular diseases (CVDs) has been extensively investigated in observational studies; however, the extent of any causal effect remains unclear, making it difficult to evaluate its clinical relevance. METHODS AND FINDINGS A phenome-wide association study (PheWAS) together with a Bayesian analysis of tree-structured phenotypic model (TreeWAS) was performed to examine disease outcomes related to genetically determined serum urate levels in 339,256 unrelated White British individuals (54% female) in the UK Biobank who were aged 40-69 years (mean age, 56.87; SD, 7.99) when recruited from 2006 to 2010. Mendelian randomization (MR) analyses were performed to replicate significant findings using various genome-wide association study (GWAS) consortia data. Sensitivity analyses were conducted to examine possible pleiotropic effects on metabolic traits of the genetic variants used as instruments for urate. PheWAS analysis, examining the association with 1,431 disease outcomes, identified 13 distinct phecodes representing 4 disease groups (inflammatory polyarthropathies, hypertensive disease, circulatory disease, and metabolic disorders) and 9 disease outcomes (gout, gouty arthropathy, pyogenic arthritis, essential hypertension, coronary atherosclerosis, ischemic heart disease, chronic ischemic heart disease, myocardial infarction, and hypercholesterolemia) that were associated with genetically determined serum urate levels after multiple testing correction (p < 3.35 × 10-4). TreeWAS analysis, examining 10,750 ICD-10 diagnostic terms, identified more sub-phenotypes of cardiovascular and cerebrovascular diseases (e.g., angina pectoris, heart failure, cerebral infarction). MR analysis successfully replicated the association with gout, hypertension, heart diseases, and blood lipid levels but indicated the existence of genetic pleiotropy. Sensitivity analyses support an inference that pleiotropic effects of genetic variants on urate and metabolic traits contribute to the observational associations with CVDs. The main limitations of this study relate to possible bias from pleiotropic effects of the considered genetic variants and possible misclassification of cases for mild disease that did not require hospitalization. CONCLUSION In this study, high serum urate levels were found to be associated with increased risk of different types of cardiac events. The finding of genetic pleiotropy indicates the existence of common upstream pathological elements influencing both urate and metabolic traits, and this may suggest new opportunities and challenges for developing drugs targeting a common mediator that would be beneficial for both the treatment of gout and the prevention of cardiovascular comorbidities.
Collapse
|
119
|
Tin A, Marten J, Halperin Kuhns VL, Li Y, Wuttke M, Kirsten H, Sieber KB, Qiu C, Gorski M, Yu Z, Giri A, Sveinbjornsson G, Li M, Chu AY, Hoppmann A, O'Connor LJ, Prins B, Nutile T, Noce D, Akiyama M, Cocca M, Ghasemi S, van der Most PJ, Horn K, Xu Y, Fuchsberger C, Sedaghat S, Afaq S, Amin N, Ärnlöv J, Bakker SJL, Bansal N, Baptista D, Bergmann S, Biggs ML, Biino G, Boerwinkle E, Bottinger EP, Boutin TS, Brumat M, Burkhardt R, Campana E, Campbell A, Campbell H, Carroll RJ, Catamo E, Chambers JC, Ciullo M, Concas MP, Coresh J, Corre T, Cusi D, Felicita SC, de Borst MH, De Grandi A, de Mutsert R, de Vries APJ, Delgado G, Demirkan A, Devuyst O, Dittrich K, Eckardt KU, Ehret G, Endlich K, Evans MK, Gansevoort RT, Gasparini P, Giedraitis V, Gieger C, Girotto G, Gögele M, Gordon SD, Gudbjartsson DF, Gudnason V, Haller T, Hamet P, Harris TB, Hayward C, Hicks AA, Hofer E, Holm H, Huang W, Hutri-Kähönen N, Hwang SJ, Ikram MA, Lewis RM, Ingelsson E, Jakobsdottir J, Jonsdottir I, Jonsson H, Joshi PK, Josyula NS, Jung B, Kähönen M, Kamatani Y, Kanai M, Kerr SM, Kiess W, Kleber ME, Koenig W, et alTin A, Marten J, Halperin Kuhns VL, Li Y, Wuttke M, Kirsten H, Sieber KB, Qiu C, Gorski M, Yu Z, Giri A, Sveinbjornsson G, Li M, Chu AY, Hoppmann A, O'Connor LJ, Prins B, Nutile T, Noce D, Akiyama M, Cocca M, Ghasemi S, van der Most PJ, Horn K, Xu Y, Fuchsberger C, Sedaghat S, Afaq S, Amin N, Ärnlöv J, Bakker SJL, Bansal N, Baptista D, Bergmann S, Biggs ML, Biino G, Boerwinkle E, Bottinger EP, Boutin TS, Brumat M, Burkhardt R, Campana E, Campbell A, Campbell H, Carroll RJ, Catamo E, Chambers JC, Ciullo M, Concas MP, Coresh J, Corre T, Cusi D, Felicita SC, de Borst MH, De Grandi A, de Mutsert R, de Vries APJ, Delgado G, Demirkan A, Devuyst O, Dittrich K, Eckardt KU, Ehret G, Endlich K, Evans MK, Gansevoort RT, Gasparini P, Giedraitis V, Gieger C, Girotto G, Gögele M, Gordon SD, Gudbjartsson DF, Gudnason V, Haller T, Hamet P, Harris TB, Hayward C, Hicks AA, Hofer E, Holm H, Huang W, Hutri-Kähönen N, Hwang SJ, Ikram MA, Lewis RM, Ingelsson E, Jakobsdottir J, Jonsdottir I, Jonsson H, Joshi PK, Josyula NS, Jung B, Kähönen M, Kamatani Y, Kanai M, Kerr SM, Kiess W, Kleber ME, Koenig W, Kooner JS, Körner A, Kovacs P, Krämer BK, Kronenberg F, Kubo M, Kühnel B, La Bianca M, Lange LA, Lehne B, Lehtimäki T, Liu J, Loeffler M, Loos RJF, Lyytikäinen LP, Magi R, Mahajan A, Martin NG, März W, Mascalzoni D, Matsuda K, Meisinger C, Meitinger T, Metspalu A, Milaneschi Y, O'Donnell CJ, Wilson OD, Gaziano JM, Mishra PP, Mohlke KL, Mononen N, Montgomery GW, Mook-Kanamori DO, Müller-Nurasyid M, Nadkarni GN, Nalls MA, Nauck M, Nikus K, Ning B, Nolte IM, Noordam R, O'Connell JR, Olafsson I, Padmanabhan S, Penninx BWJH, Perls T, Peters A, Pirastu M, Pirastu N, Pistis G, Polasek O, Ponte B, Porteous DJ, Poulain T, Preuss MH, Rabelink TJ, Raffield LM, Raitakari OT, Rettig R, Rheinberger M, Rice KM, Rizzi F, Robino A, Rudan I, Krajcoviechova A, Cifkova R, Rueedi R, Ruggiero D, Ryan KA, Saba Y, Salvi E, Schmidt H, Schmidt R, Shaffer CM, Smith AV, Smith BH, Spracklen CN, Strauch K, Stumvoll M, Sulem P, Tajuddin SM, Teren A, Thiery J, Thio CHL, Thorsteinsdottir U, Toniolo D, Tönjes A, Tremblay J, Uitterlinden AG, Vaccargiu S, van der Harst P, van Duijn CM, Verweij N, Völker U, Vollenweider P, Waeber G, Waldenberger M, Whitfield JB, Wild SH, Wilson JF, Yang Q, Zhang W, Zonderman AB, Bochud M, Wilson JG, Pendergrass SA, Ho K, Parsa A, Pramstaller PP, Psaty BM, Böger CA, Snieder H, Butterworth AS, Okada Y, Edwards TL, Stefansson K, Susztak K, Scholz M, Heid IM, Hung AM, Teumer A, Pattaro C, Woodward OM, Vitart V, Köttgen A. Target genes, variants, tissues and transcriptional pathways influencing human serum urate levels. Nat Genet 2019; 51:1459-1474. [PMID: 31578528 PMCID: PMC6858555 DOI: 10.1038/s41588-019-0504-x] [Show More Authors] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022]
Abstract
Elevated serum urate levels cause gout and correlate with cardiometabolic diseases via poorly understood mechanisms. We performed a trans-ancestry genome-wide association study of serum urate in 457,690 individuals, identifying 183 loci (147 previously unknown) that improve the prediction of gout in an independent cohort of 334,880 individuals. Serum urate showed significant genetic correlations with many cardiometabolic traits, with genetic causality analyses supporting a substantial role for pleiotropy. Enrichment analysis, fine-mapping of urate-associated loci and colocalization with gene expression in 47 tissues implicated the kidney and liver as the main target organs and prioritized potentially causal genes and variants, including the transcriptional master regulators in the liver and kidney, HNF1A and HNF4A. Experimental validation showed that HNF4A transactivated the promoter of ABCG2, encoding a major urate transporter, in kidney cells, and that HNF4A p.Thr139Ile is a functional variant. Transcriptional coregulation within and across organs may be a general mechanism underlying the observed pleiotropy between urate and cardiometabolic traits.
Collapse
Affiliation(s)
- Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Welch Centre for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA.
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Yong Li
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Karsten B Sieber
- Target Sciences-Genetics, GlaxoSmithKline, Collegeville, PA, USA
| | - Chengxiang Qiu
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Mathias Gorski
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Zhi Yu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ayush Giri
- Division of Quantitative Sciences, Department of Obstetrics & Gynecology, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | | | - Man Li
- Department of Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, UT, USA
| | | | - Anselm Hoppmann
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Luke J O'Connor
- Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bram Prins
- Strangeways Research Laboratory, University of Cambridge, Cambridge, UK
| | - Teresa Nutile
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
| | - Damia Noce
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Massimiliano Cocca
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Sahar Ghasemi
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Katrin Horn
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Yizhe Xu
- Department of Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, UT, USA
| | | | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Saima Afaq
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Institute of Public Health & Social Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Division of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nisha Bansal
- Division of Nephrology, University of Washington, Seattle, WA, USA
- Kidney Research Institute, University of Washington, Seattle, WA, USA
| | | | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Mary L Biggs
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Pavia, Italy
| | - Eric Boerwinkle
- Human Genetics Centre, University of Texas Health Science Centre, Houston, TX, USA
| | - Erwin P Bottinger
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thibaud S Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Marco Brumat
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Ralph Burkhardt
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Eric Campana
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Robert J Carroll
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eulalia Catamo
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Cardiology, Ealing Hospital, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Marina Ciullo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Tanguy Corre
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Daniele Cusi
- Institute of Biomedical Technologies, Italy National Research Council, Milano, Italy
- Bio4Dreams, Milano, Italy
| | | | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Aiko P J de Vries
- Section of Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Graciela Delgado
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ayşe Demirkan
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Katalin Dittrich
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Georg Ehret
- Cardiology, Geneva University Hospitals, Geneva, Switzerland
| | - Karlhans Endlich
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Paolo Gasparini
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Giorgia Girotto
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Martin Gögele
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Toomas Haller
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Pavel Hamet
- Montreal University Hospital Research Centre, Centre Hospitalier de lUniversité de Montréal, Montreal, Quebec, Canada
- Medpharmgene, Montreal, Quebec, Canada
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Andrew A Hicks
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Hilma Holm
- deCODE Genetics, Amgen Inc., Reykjavik, Iceland
| | - Wei Huang
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Centre, Shanghai, China
- Shanghai Industrial Technology Institute, Shanghai, China
| | - Nina Hutri-Kähönen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Pediatrics, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- The Centre for Population Studies, National Heart, Lung, and Blood Institute, Framingham, MA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Raychel M Lewis
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Molecular Epidemiology and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Johanna Jakobsdottir
- Icelandic Heart Association, Kópavogur, Iceland
- The Centre of Public Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Helgi Jonsson
- Landspitalinn University Hospital, Reykjavík, Iceland
- University of Iceland, Reykjavík, Iceland
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Navya Shilpa Josyula
- Geisinger Research, Biomedical and Translational Informatics Institute, Rockville, MD, USA
| | - Bettina Jung
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Shona M Kerr
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Wieland Kiess
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Marcus E Kleber
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Biostatistics, University of Ulm, Ulm, Germany
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, 323 School of Public Health, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Antje Körner
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Integrated Research and Treatment Centre Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michiaki Kubo
- RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
| | - Martina La Bianca
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jun Liu
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Reedik Magi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Winfried März
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | | | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Christa Meisinger
- Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-Universität München at UNIKA-T Augsburg, Augsburg, Germany
| | - Thomas Meitinger
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Christopher J O'Donnell
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Otis D Wilson
- Vanderbilt University Medical Centre, Division of Nephrology & Hypertension, Nashville, TN, USA
| | - J Michael Gaziano
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Centre, Leiden, the Netherlands
| | - Martina Müller-Nurasyid
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Matthias Nauck
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
- Department of Cardiology, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Boting Ning
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali University Hospital, Reykjavik, Iceland
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Thomas Perls
- Department of Medicine, Geriatrics Section, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Mario Pirastu
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Sassari, Sassari, Italy
| | - Nicola Pirastu
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Giorgio Pistis
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
- Gen-info Ltd, Zagreb, Croatia
| | - Belen Ponte
- Nephrology Service, Department of Specialties in Internal Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Tanja Poulain
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ton J Rabelink
- Section of Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
- Einthoven Laboratory of Experimental Vascular Research, Leiden University Medical Centre, Leiden, the Netherlands
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Rainer Rettig
- Institute of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Myriam Rheinberger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Federica Rizzi
- Department of Health Sciences, University of Milan, Milano, Italy
- ePhood Scientific Unit, ePhood SRL, Milano, Italy
| | - Antonietta Robino
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Alena Krajcoviechova
- Center for Cardiovascular Prevention, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Thomayer Hospital, Prague, Czech Republic
| | - Renata Cifkova
- Center for Cardiovascular Prevention, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Medicine II, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Kathleen A Ryan
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yasaman Saba
- Molecular Biology and Biochemistry, Gottfried Schatz Research Centre for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Erika Salvi
- Department of Health Sciences, University of Milan, Milano, Italy
- Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Christian M Shaffer
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Albert V Smith
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | | | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Stumvoll
- Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
| | | | - Salman M Tajuddin
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Andrej Teren
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Heart Centre Leipzig, Leipzig, Germany
| | - Joachim Thiery
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Chris H L Thio
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | | | - Anke Tönjes
- Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Johanne Tremblay
- Montreal University Hospital Research Centre, Centre Hospitalier de lUniversité de Montréal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Simona Vaccargiu
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Sassari, Sassari, Italy
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Durrer Centre for Cardiovascular Research, the Netherlands Heart Institute, Utrecht, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Genomics plc, Oxford, UK
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Peter Vollenweider
- Internal Medicine, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Gerard Waeber
- Internal Medicine, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - John B Whitfield
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sarah H Wild
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - James F Wilson
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London, UK
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Murielle Bochud
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Centre, Jackson, MS, USA
| | - Sarah A Pendergrass
- Geisinger Research, Biomedical and Translational Informatics Institute, Danville, PA, USA
| | - Kevin Ho
- Kidney Health Research Institute, Geisinger, Danville, PA, USA
- Department of Nephrology, Geisinger, Danville, PA, USA
| | - Afshin Parsa
- Division of Kidney, Urologic and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, Department of Epidemiology, Department of Health Service, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Nephrology and Rheumatology, Kliniken Südostbayern AG, Traunstein, Germany
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Adam S Butterworth
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Osaka, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Centre, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | | | - Katalin Susztak
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Adriana M Hung
- Vanderbilt University Medical Centre, Division of Nephrology & Hypertension, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | | | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany.
| |
Collapse
|
120
|
Kawamura Y, Nakaoka H, Nakayama A, Okada Y, Yamamoto K, Higashino T, Sakiyama M, Shimizu T, Ooyama H, Ooyama K, Nagase M, Hidaka Y, Shirahama Y, Hosomichi K, Nishida Y, Shimoshikiryo I, Hishida A, Katsuura-Kamano S, Shimizu S, Kawaguchi M, Uemura H, Ibusuki R, Hara M, Naito M, Takao M, Nakajima M, Iwasawa S, Nakashima H, Ohnaka K, Nakamura T, Stiburkova B, Merriman TR, Nakatochi M, Ichihara S, Yokota M, Takada T, Saitoh T, Kamatani Y, Takahashi A, Arisawa K, Takezaki T, Tanaka K, Wakai K, Kubo M, Hosoya T, Ichida K, Inoue I, Shinomiya N, Matsuo H. Genome-wide association study revealed novel loci which aggravate asymptomatic hyperuricaemia into gout. Ann Rheum Dis 2019; 78:1430-1437. [PMID: 31289104 PMCID: PMC6788923 DOI: 10.1136/annrheumdis-2019-215521] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The first ever genome-wide association study (GWAS) of clinically defined gout cases and asymptomatic hyperuricaemia (AHUA) controls was performed to identify novel gout loci that aggravate AHUA into gout. METHODS We carried out a GWAS of 945 clinically defined gout cases and 1003 AHUA controls followed by 2 replication studies. In total, 2860 gout cases and 3149 AHUA controls (all Japanese men) were analysed. We also compared the ORs for each locus in the present GWAS (gout vs AHUA) with those in the previous GWAS (gout vs normouricaemia). RESULTS This new approach enabled us to identify two novel gout loci (rs7927466 of CNTN5 and rs9952962 of MIR302F) and one suggestive locus (rs12980365 of ZNF724) at the genome-wide significance level (p<5.0×10-8). The present study also identified the loci of ABCG2, ALDH2 and SLC2A9. One of them, rs671 of ALDH2, was identified as a gout locus by GWAS for the first time. Comparing ORs for each locus in the present versus the previous GWAS revealed three 'gout vs AHUA GWAS'-specific loci (CNTN5, MIR302F and ZNF724) to be clearly associated with mechanisms of gout development which distinctly differ from the known gout risk loci that basically elevate serum uric acid level. CONCLUSIONS This meta-analysis is the first to reveal the loci associated with crystal-induced inflammation, the last step in gout development that aggravates AHUA into gout. Our findings should help to elucidate the molecular mechanisms of gout development and assist the prevention of gout attacks in high-risk AHUA individuals.
Collapse
Affiliation(s)
- Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
- Department of General Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hirofumi Nakaoka
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
- Medical Squadron, Air Base Group, Western Aircraft Control and Warning Wing, Japan Air Self-Defense Force, Kasuga, Fukuoka, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Toshihide Higashino
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Masayuki Sakiyama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
- Department of Defense Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Toru Shimizu
- Midorigaoka Hospital, Takatsuki, Osaka, Japan
- Kyoto Industrial Health Association, Kyoto, Japan
| | | | | | | | | | - Yuko Shirahama
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Ippei Shimoshikiryo
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Sakurako Katsuura-Kamano
- Department of Preventive Medicine, Institute of Health Biosciences, the University of Tokushima Graduate School, Tokushima, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Makoto Kawaguchi
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
- Department of Urology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hirokazu Uemura
- Department of Preventive Medicine, Institute of Health Biosciences, the University of Tokushima Graduate School, Tokushima, Japan
| | - Rie Ibusuki
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Oral Epidemiology, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Mikiya Takao
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Mayuko Nakajima
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Satoko Iwasawa
- Department of Preventive Medicine and Public Health, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroshi Nakashima
- Department of Preventive Medicine and Public Health, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Keizo Ohnaka
- Department of Geriatric Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Nakamura
- Laboratory for Mathematics, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Blanka Stiburkova
- Institute of Rheumatology, Prague, Czech Republic
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tony R Merriman
- Department of Biochemisty, University of Otago, Dunedin, New Zealand
| | - Masahiro Nakatochi
- Data Science Division, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| | - Mitsuhiro Yokota
- Department of Genome Science, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| | - Tappei Takada
- Department of Pharmacy, the University of Tokyo Hospital, Tokyo, Japan
| | - Tatsuya Saitoh
- Laboratory of Bioresponse Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
- Division of Inflammation Biology, Institute for Enzyme Research, Tokushima University, Tokushima, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Genomic Medicine, Research Institute, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Kokichi Arisawa
- Department of Preventive Medicine, Institute of Health Biosciences, the University of Tokushima Graduate School, Tokushima, Japan
| | - Toshiro Takezaki
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Keitaro Tanaka
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Tatsuo Hosoya
- Division of Kidney and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Department of Pathophysiology and Therapy in Chronic Kidney Disease, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Kimiyoshi Ichida
- Division of Kidney and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Ituro Inoue
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
121
|
van der Wijst J, Belge H, Bindels RJM, Devuyst O. Learning Physiology From Inherited Kidney Disorders. Physiol Rev 2019; 99:1575-1653. [PMID: 31215303 DOI: 10.1152/physrev.00008.2018] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The identification of genes causing inherited kidney diseases yielded crucial insights in the molecular basis of disease and improved our understanding of physiological processes that operate in the kidney. Monogenic kidney disorders are caused by mutations in genes coding for a large variety of proteins including receptors, channels and transporters, enzymes, transcription factors, and structural components, operating in specialized cell types that perform highly regulated homeostatic functions. Common variants in some of these genes are also associated with complex traits, as evidenced by genome-wide association studies in the general population. In this review, we discuss how the molecular genetics of inherited disorders affecting different tubular segments of the nephron improved our understanding of various transport processes and of their involvement in homeostasis, while providing novel therapeutic targets. These include inherited disorders causing a dysfunction of the proximal tubule (renal Fanconi syndrome), with emphasis on epithelial differentiation and receptor-mediated endocytosis, or affecting the reabsorption of glucose, the handling of uric acid, and the reabsorption of sodium, calcium, and magnesium along the kidney tubule.
Collapse
Affiliation(s)
- Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Hendrica Belge
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Devuyst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; Institute of Physiology, University of Zurich , Zurich , Switzerland ; and Division of Nephrology, Institute of Experimental and Clinical Research (IREC), Medical School, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
122
|
The association between genotypes of urate transporter-1, Serum uric acid, and mortality in the community-based population: the Yamagata (Takahata) Study. Clin Exp Nephrol 2019; 23:1357-1363. [PMID: 31478104 DOI: 10.1007/s10157-019-01781-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 08/12/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND The urate transporter-1 (URAT1) is crucial in developing hyperuricemia via reabsorption of uric acid in renal tubules, and its function is regulated by several single nucleotide polymorphisms (SNPs) within SLC22A12 gene encoding URAT1. This study investigated whether the genetic predisposition of URAT1 is associated with the mortality in general population. METHODS This study enrolled 1596 participants (male 45%, mean age 61 years) who registered at local health checkup in Takahata, Japan, and the association between the rs505802 genotypes in SLC22A12 gene and the 7-year mortality, was examined. RESULTS The serum uric acid levels (mean ± SD) at baseline in the subjects with GG and AG + AA genotypes of rs505802 were 5.1 ± 1.3 mg/dL and 5.0 ± 1.5 mg/dL, respectively. Kaplan-Meier analysis revealed that the mortality was nonsignificantly higher in the subjects with GG genotype than in those with AG + AA genotype (P = 0.09). Cox proportional hazard model adjusted with age, gender, renal function, comorbidities, and other possible confounders, demonstrated that the GG genotype was significantly associated with the mortality [hazard ratio (HR) 2.23, 95% confidence interval (CI) 1.05-4.85, (vs. AG + AA genotype)]. Furthermore, adjustment with serum uric acid levels, along with aforementioned confounders retained the significant association (HR 2.26, 95% CI 1.05-4.85). CONCLUSIONS This study revealed that the genetic predisposition of URAT1 was independently associated with mortality in the Japanese community-based population. This association might be due to the mechanism independent of serum uric acid levels.
Collapse
|
123
|
Lee YH, Song GG. Uric acid level, gout and bone mineral density: A Mendelian randomization study. Eur J Clin Invest 2019; 49:e13156. [PMID: 31294819 DOI: 10.1111/eci.13156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/28/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This study aimed to examine whether the uric acid level or gout is causally associated with bone mineral density (BMD). METHOD We performed a two-sample Mendelian randomization (MR) analysis. The statistics dataset, we used was from a meta-analysis of genome-wide association studies (GWASs) on uric acid levels from 14 studies with a total of 28 141 participants of European descent, and the dataset for gout from the United Kingdom (UK) Biobank (4807 cases and 3 32 352 controls). We further used the summary statistics dataset of a GWAS on lumbar spine and femur neck (FN) BMDs of individuals of European ancestry (up to 32 735). RESULTS The instrumental variables (IVs) selected were six single nucleotide polymorphisms (SNPs) from the uric acid level GWAS data and 19 SNPs from the gout GWAS data. The inverse-variance weighted (IVW) method yielded no evidence to support a causal association between the uric acid level or gout and lumbar spine BMD (β = -.002, standard error (SE) = 0.035, P = .951; β = -.700, SE = 0.672, P = .297). MR-Egger regression revealed no causality between uric acid level, gout and lumbar spine. Similarly, the weighted median approach provided no evidence of causality between uric acid level, gout and lumbar spine BMD. The MR results on FN BMD showed similar patterns with those of the lumbar spine BMD. CONCLUSIONS Mendelian randomization analysis did not support a causal association between uric acid level, gout and lumbar spine or FN BMD.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
124
|
Kia DA, Noyce AJ, White J, Speed D, Nicolas A, Burgess S, Lawlor DA, Davey Smith G, Singleton A, Nalls MA, Sofat R, Wood NW. Mendelian randomization study shows no causal relationship between circulating urate levels and Parkinson's disease. Ann Neurol 2019; 84:191-199. [PMID: 30014513 PMCID: PMC6481555 DOI: 10.1002/ana.25294] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/30/2018] [Accepted: 06/04/2018] [Indexed: 02/02/2023]
Abstract
Objective Observational studies have shown that increased plasma urate is associated with lower risk of Parkinson’s disease (PD), but these studies were not designed to test causality. If a causal relationship exists, then modulating plasma urate levels could be a potential preventive avenue for PD. We used a large two-sample Mendelian randomization (MR) design to assess for a causal relationship between plasma urate and PD risk. Methods We used a genetic instrument consisting of 31 independent loci for plasma urate on a case-control genome-wide association study data set, which included 13,708 PD cases and 95,282 controls. Individual effect estimates for each SNP were combined using the inverse-variance weighted (IVW) method. Two additional methods, MR-Egger and a penalized weighted median (PWM)-based approach, were used to assess potential bias attributed to pleiotropy or invalid instruments. Results We found no evidence for a causal relationship between urate and PD, with an effect estimate from the IVW method of odds ratio (OR) 1.03 (95% confidence interval [CI], 0.88–1.20) per 1-standard-deviation increase in plasma urate levels. MR Egger and PWM analyses yielded similar estimates (OR, 0.99 [95% CI, 0.83–1.17] and 0.99 [95% CI, 0.86−1.14], respectively). Interpretation We did not find evidence for a linear causal protective effect by urate on PD risk. The associations observed in previous observational studies may be, in part, attributed to confounding or reverse causality. In the context of the present findings, strategies to elevate circulating urate levels may not reduce overall PD risk.
Collapse
Affiliation(s)
- Demis A Kia
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | - Alastair J Noyce
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom.,Preventive Neurology Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom
| | - Jon White
- UCL Genetics Institute, University College, London, United Kingdom
| | - Doug Speed
- UCL Genetics Institute, University College, London, United Kingdom
| | - Aude Nicolas
- Laboratory for Neurogenetics, National Institutes for Health, Bethesda, MD
| | | | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom.,Population Health Science, Bristol Medical School of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom.,Population Health Science, Bristol Medical School of Bristol, Bristol, United Kingdom
| | - Andrew Singleton
- Laboratory for Neurogenetics, National Institutes for Health, Bethesda, MD
| | - Mike A Nalls
- Laboratory for Neurogenetics, National Institutes for Health, Bethesda, MD.,Data Tecnica International, Glen Echo, MD
| | - Reecha Sofat
- Institute of Health Informatics, University College London, London, United Kingdom
| | - Nicholas W Wood
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| |
Collapse
|
125
|
Major TJ, Dalbeth N, Stahl EA, Merriman TR. An update on the genetics of hyperuricaemia and gout. Nat Rev Rheumatol 2019; 14:341-353. [PMID: 29740155 DOI: 10.1038/s41584-018-0004-x] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A central aspect of the pathogenesis of gout is elevated urate concentrations, which lead to the formation of monosodium urate crystals. The clinical features of gout result from an individual's immune response to these deposited crystals. Genome-wide association studies (GWAS) have confirmed the importance of urate excretion in the control of serum urate levels and the risk of gout and have identified the kidneys, the gut and the liver as sites of urate regulation. The genetic contribution to the progression from hyperuricaemia to gout remains relatively poorly understood, although genes encoding proteins that are involved in the NLRP3 (NOD-, LRR- and pyrin domain-containing 3) inflammasome pathway play a part. Genome-wide and targeted sequencing is beginning to identify uncommon population-specific variants that are associated with urate levels and gout. Mendelian randomization studies using urate-associated genetic variants as unconfounded surrogates for lifelong urate exposure have not supported claims that urate is causal for metabolic conditions that are comorbidities of hyperuricaemia and gout. Genetic studies have also identified genetic variants that predict responsiveness to therapies (for example, urate-lowering drugs) for treatment of hyperuricaemia. Future research should focus on large GWAS (that include asymptomatic hyperuricaemic individuals) and on increasing the use of whole-genome sequencing data to identify uncommon genetic variants with increased penetrance that might provide opportunities for clinical translation.
Collapse
Affiliation(s)
- Tanya J Major
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Eli A Stahl
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
126
|
Ellul S, Wake M, Clifford SA, Lange K, Würtz P, Juonala M, Dwyer T, Carlin JB, Burgner DP, Saffery R. Metabolomics: population epidemiology and concordance in Australian children aged 11-12 years and their parents. BMJ Open 2019; 9:106-117. [PMID: 31273021 PMCID: PMC6624050 DOI: 10.1136/bmjopen-2017-020900] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Nuclear magnetic resonance (NMR) metabolomics is high throughput and cost-effective, with the potential to improve the understanding of disease and risk. We examine the circulating metabolic profile by quantitative NMR metabolomics of a sample of Australian 11-12 year olds children and their parents, describe differences by age and sex, and explore the correlation of metabolites in parent-child dyads. DESIGN The population-based cross-sectional Child Health CheckPoint study nested within the Longitudinal Study of Australian Children. SETTING Blood samples collected from CheckPoint participants at assessment centres in seven Australian cities and eight regional towns; February 2015-March 2016. PARTICIPANTS 1180 children and 1325 parents provided a blood sample and had metabolomics data available. This included 1133 parent-child dyads (518 mother-daughter, 469 mother-son, 68 father-daughter and 78 father-son). OUTCOME MEASURES 228 metabolic measures were obtained for each participant. We focused on 74 biomarkers including amino acid species, lipoprotein subclass measures, lipids, fatty acids, measures related to fatty acid saturation, and composite markers of inflammation and energy homeostasis. RESULTS We identified differences in the concentration of specific metabolites between childhood and adulthood and in metabolic profiles in children and adults by sex. In general, metabolite concentrations were higher in adults than children and sex differences were larger in adults than in children. Positive correlations were observed for the majority of metabolites including isoleucine (CC 0.33, 95% CI 0.27 to 0.38), total cholesterol (CC 0.30, 95% CI 0.24 to 0.35) and omega 6 fatty acids (CC 0.28, 95% CI 0.23 to 0.34) in parent-child comparisons. CONCLUSIONS We describe the serum metabolite profiles from mid-childhood and adulthood in a population-based sample, together with a parent-child concordance. Differences in profiles by age and sex were observed. These data will be informative for investigation of the childhood origins of adult non-communicable diseases and for comparative studies in other populations.
Collapse
Affiliation(s)
- Susan Ellul
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
| | - Melissa Wake
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics and The Liggins Institute, University of Auckland, Auckland, New Zealand
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan A Clifford
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Katherine Lange
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Würtz
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Nightingale Health Ltd., Helsinki, Finland
| | - Markus Juonala
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Terence Dwyer
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- The George Institute for Global Health, Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
- Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - John B Carlin
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - David P Burgner
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Richard Saffery
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
127
|
Rivera-Paredez B, Macías-Kauffer L, Fernandez-Lopez JC, Villalobos-Comparán M, Martinez-Aguilar MM, de la Cruz-Montoya A, Ramírez-Salazar EG, Villamil-Ramírez H, Quiterio M, Ramírez-Palacios P, Romero-Hidalgo S, Villarreal-Molina MT, Denova-Gutiérrez E, Flores YN, Canizales-Quinteros S, Salmerón J, Velázquez-Cruz R. Influence of Genetic and Non-Genetic Risk Factors for Serum Uric Acid Levels and Hyperuricemia in Mexicans. Nutrients 2019; 11:nu11061336. [PMID: 31207883 PMCID: PMC6627998 DOI: 10.3390/nu11061336] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/29/2019] [Accepted: 06/10/2019] [Indexed: 12/21/2022] Open
Abstract
Risk of hyperuricemia is modified by genetic and environmental factors. Our aim was to identify factors associated with serum uric acid levels and hyperuricemia in Mexicans. A pilot Genome-wide association study GWAS was performed in a subgroup of participants (n = 411) from the Health Workers Cohort Study (HWCS). Single nucleotide polymorphisms (SNPs) associated with serum uric acid levels were validated in all the HWCS participants (n = 1939) and replicated in independent children (n = 1080) and adult (n = 1073) case-control studies. The meta-analysis of the whole HWCS and replication samples identified three SLC2A9 SNPs: rs1014290 (p = 2.3 × 10−64), rs3775948 (p = 8.2 × 10−64) and rs11722228 (p = 1.1 × 10−17); and an ABCG2 missense SNP, rs2231142 (p = 1.0 × 10−18). Among the non-genetic factors identified, the visceral adiposity index, smoking, the metabolic syndrome and its components (waist circumference, blood pressure, glucose and hyperlipidemia) were associated with increased serum uric acid levels and hyperuricemia (p < 0.05). Among the female HWCS participants, the odds ratio for hyperuricemia was 1.24 (95% CI, 1.01–1.53) per unit increase in soft drink consumption. As reported in other studies, our findings indicate that diet, adiposity and genetic variation contribute to the elevated prevalence of hyperuricemia in Mexico.
Collapse
Affiliation(s)
- Berenice Rivera-Paredez
- Centro de Investigación en Políticas, Población y Salud de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Luis Macías-Kauffer
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México 14610, Mexico.
| | | | | | - Mayeli M Martinez-Aguilar
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México 14610, Mexico.
| | - Aldo de la Cruz-Montoya
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México 14610, Mexico.
| | - Eric G Ramírez-Salazar
- Consejo Nacional de Ciencia y Tecnología (CONACYT)-Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico.
| | - Hugo Villamil-Ramírez
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México 14610, Mexico.
| | - Manuel Quiterio
- Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública. Cuernavaca, Morelos 62100, Mexico.
| | - Paula Ramírez-Palacios
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Instituto Mexicano del Seguro Social (IMSS), Cuernavaca, Morelos 62000, Mexico.
| | - Sandra Romero-Hidalgo
- Consorcio Genómica Computacional, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico.
| | | | - Edgar Denova-Gutiérrez
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, Morelos 62100, Mexico.
| | - Yvonne N Flores
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Instituto Mexicano del Seguro Social (IMSS), Cuernavaca, Morelos 62000, Mexico.
- UCLA Department of Health Policy and Management, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA 90001, USA.
| | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México 14610, Mexico.
| | - Jorge Salmerón
- Centro de Investigación en Políticas, Población y Salud de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México 14610, Mexico.
| |
Collapse
|
128
|
Dual actions of norathyriol as a new candidate hypouricaemic agent: uricosuric effects and xanthine oxidase inhibition. Eur J Pharmacol 2019; 853:371-380. [DOI: 10.1016/j.ejphar.2019.04.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 01/16/2023]
|
129
|
Cameron-Christie S, Wolock CJ, Groopman E, Petrovski S, Kamalakaran S, Povysil G, Vitsios D, Zhang M, Fleckner J, March RE, Gelfman S, Marasa M, Li Y, Sanna-Cherchi S, Kiryluk K, Allen AS, Fellström BC, Haefliger C, Platt A, Goldstein DB, Gharavi AG. Exome-Based Rare-Variant Analyses in CKD. J Am Soc Nephrol 2019; 30:1109-1122. [PMID: 31085678 DOI: 10.1681/asn.2018090909] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 03/06/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Studies have identified many common genetic associations that influence renal function and all-cause CKD, but these explain only a small fraction of variance in these traits. The contribution of rare variants has not been systematically examined. METHODS We performed exome sequencing of 3150 individuals, who collectively encompassed diverse CKD subtypes, and 9563 controls. To detect causal genes and evaluate the contribution of rare variants we used collapsing analysis, in which we compared the proportion of cases and controls carrying rare variants per gene. RESULTS The analyses captured five established monogenic causes of CKD: variants in PKD1, PKD2, and COL4A5 achieved study-wide significance, and we observed suggestive case enrichment for COL4A4 and COL4A3. Beyond known disease-associated genes, collapsing analyses incorporating regional variant intolerance identified suggestive dominant signals in CPT2 and several other candidate genes. Biallelic mutations in CPT2 cause carnitine palmitoyltransferase II deficiency, sometimes associated with rhabdomyolysis and acute renal injury. Genetic modifier analysis among cases with APOL1 risk genotypes identified a suggestive signal in AHDC1, implicated in Xia-Gibbs syndrome, which involves intellectual disability and other features. On the basis of the observed distribution of rare variants, we estimate that a two- to three-fold larger cohort would provide 80% power to implicate new genes for all-cause CKD. CONCLUSIONS This study demonstrates that rare-variant collapsing analyses can validate known genes and identify candidate genes and modifiers for kidney disease. In so doing, these findings provide a motivation for larger-scale investigation of rare-variant risk contributions across major clinical CKD categories.
Collapse
Affiliation(s)
- Sophia Cameron-Christie
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK
| | | | - Emily Groopman
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Slavé Petrovski
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK
| | | | - Gundula Povysil
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK.,Institute for Genomic Medicine, Columbia University Medical Center, New York, New York
| | - Dimitrios Vitsios
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK
| | - Mengqi Zhang
- Institute for Genomic Medicine, Columbia University Medical Center, New York, New York.,Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina; and
| | - Jan Fleckner
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK
| | - Ruth E March
- Precision Medicine, R&D Oncology, AstraZeneca, Cambridge, UK
| | | | - Maddalena Marasa
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Yifu Li
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Andrew S Allen
- Institute for Genomic Medicine, Columbia University Medical Center, New York, New York.,Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina; and
| | - Bengt C Fellström
- Department of Medical Sciences, Uppsala University Hospital, Uppsala, Sweden; and
| | - Carolina Haefliger
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK
| | - Adam Platt
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK;
| | - David B Goldstein
- AstraZeneca Centre for Genomics Research, Discovery Sciences, R&D BioPharmaceuticals, AstraZeneca, Cambridge, UK; .,Department of Genetics and Development and.,Institute for Genomic Medicine, Columbia University Medical Center, New York, New York
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York; .,Institute for Genomic Medicine, Columbia University Medical Center, New York, New York
| |
Collapse
|
130
|
Han X, Yang J, Li D, Guo Z. Overexpression of Uric Acid Transporter SLC2A9 Inhibits Proliferation of Hepatocellular Carcinoma Cells. Oncol Res 2019. [PMID: 29523220 PMCID: PMC7848443 DOI: 10.3727/096504018x15199489058224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-associated mortality worldwide. Although the mechanisms of HCC progression are not well understood, recent studies demonstrated the potential contribution of uric acid transporter SLC2A9 to tumor suppression. However, the roles and underlying mechanisms are still unknown. We aimed to study the roles and mechanisms of SLC2A9 in HCC. The present study showed that SLC2A9 expression was decreased in human HCC tissues and cell lines. In addition, overexpression of SLC2A9 inhibited HCC cell proliferation. SCL2A9 induced HCC cell apoptosis by inhibiting the expression of caspase 3. Our study also revealed that upregulation of SLC2A9 reduced intracellular reactive oxygen species (ROS) accumulation. Furthermore, SLC2A9 increased the mRNA and protein expression of tumor suppressor p53 in HCC cells. Probenecid inhibits SLC2A9-mediated uric acid transport, which promotes cell proliferation, inhibits cell apoptosis, induces intracellular ROS, and decreases the expression of p53 in HCC cells. Therefore, the present study demonstrated that SLC2A9 may be a novel tumor suppressor gene and a potential therapeutic target in HCC.
Collapse
Affiliation(s)
- Xiaoying Han
- Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, P.R. China
| | - Jing Yang
- Department of Oncology, Xuzhou City Hospital of Traditional Chinese Medicine, Xuzhou, P.R. China
| | - Dong Li
- Department of Oncology, Xuzhou Central Hospital, Xuzhou, Jiangsu, P.R. China
| | - Zewei Guo
- Department of Internal Medicine, Huangshan Traditional Chinese Medicine, Huangshan, Anhui, P.R. China
| |
Collapse
|
131
|
Toyoda Y, Mančíková A, Krylov V, Morimoto K, Pavelcová K, Bohatá J, Pavelka K, Pavlíková M, Suzuki H, Matsuo H, Takada T, Stiburkova B. Functional Characterization of Clinically-Relevant Rare Variants in ABCG2 Identified in a Gout and Hyperuricemia Cohort. Cells 2019; 8:E363. [PMID: 31003562 PMCID: PMC6523779 DOI: 10.3390/cells8040363] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
ATP-binding cassette subfamily G member 2 (ABCG2) is a physiologically important urate transporter. Accumulating evidence demonstrates that congenital dysfunction of ABCG2 is an important genetic risk factor in gout and hyperuricemia; recent studies suggest the clinical significance of both common and rare variants of ABCG2. However, the effects of rare variants of ABCG2 on the risk of such diseases are not fully understood. Here, using a cohort of 250 Czech individuals of European descent (68 primary hyperuricemia patients and 182 primary gout patients), we examined exonic non-synonymous variants of ABCG2. Based on the results of direct sequencing and database information, we experimentally characterized nine rare variants of ABCG2: R147W (rs372192400), T153M (rs753759474), F373C (rs752626614), T421A (rs199854112), T434M (rs769734146), S476P (not annotated), S572R (rs200894058), D620N (rs34783571), and a three-base deletion K360del (rs750972998). Functional analyses of these rare variants revealed a deficiency in the plasma membrane localization of R147W and S572R, lower levels of cellular proteins of T153M and F373C, and null urate uptake function of T434M and S476P. Accordingly, we newly identified six rare variants of ABCG2 that showed lower or null function. Our findings contribute to deepening the understanding of ABCG2-related gout/hyperuricemia risk and the biochemical characteristics of the ABCG2 protein.
Collapse
Affiliation(s)
- Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan.
| | - Andrea Mančíková
- Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague 2, Czech Republic.
| | - Vladimír Krylov
- Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague 2, Czech Republic.
| | - Keito Morimoto
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan.
| | | | - Jana Bohatá
- Institute of Rheumatology, 128 50 Prague 2, Czech Republic.
| | - Karel Pavelka
- Institute of Rheumatology, 128 50 Prague 2, Czech Republic.
| | - Markéta Pavlíková
- Department of Probability and Mathematical Statistics, Faculty of Mathematics and Physics, Charles University, 121 16 Prague 2, Czech Republic.
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan.
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama 359-8513, Japan.
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan.
| | - Blanka Stiburkova
- Institute of Rheumatology, 128 50 Prague 2, Czech Republic.
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, 121 08 Prague 2, Czech Republic.
| |
Collapse
|
132
|
Tanikawa C, Kamatani Y, Terao C, Usami M, Takahashi A, Momozawa Y, Suzuki K, Ogishima S, Shimizu A, Satoh M, Matsuo K, Mikami H, Naito M, Wakai K, Yamaji T, Sawada N, Iwasaki M, Tsugane S, Kohri K, Yu ASL, Yasui T, Murakami Y, Kubo M, Matsuda K. Novel Risk Loci Identified in a Genome-Wide Association Study of Urolithiasis in a Japanese Population. J Am Soc Nephrol 2019; 30:855-864. [PMID: 30975718 DOI: 10.1681/asn.2018090942] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/08/2019] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND A family history of urolithiasis is associated with a more than doubling of urolithiasis risk, and a twin study estimating 56% heritability of the condition suggests a pivotal role for host genetic factors. However, previous genome-wide association studies (GWAS) have identified only six risk-related loci. METHODS To identify novel urolithiasis-related loci in the Japanese population, we performed a large-scale GWAS of 11,130 cases and 187,639 controls, followed by a replication analysis of 2289 cases and 3817 controls. Diagnosis of urolithiasis was confirmed either by a clinician or using medical records or self-report. We also assessed the association of urolithiasis loci with 16 quantitative traits, including metabolic, kidney-related, and electrolyte traits (such as body mass index, lipid storage, eGFR, serum uric acid, and serum calcium), using up to 160,000 samples from BioBank Japan. RESULTS The analysis identified 14 significant loci, including nine novel loci. Ten regions showed a significant association with at least one quantitative trait, including metabolic, kidney-related, and electrolyte traits, suggesting a common genetic basis for urolithiasis and these quantitative traits. Four novel loci were related to metabolic traits, obesity, hypertriglyceridemia, or hyperuricemia. The remaining ten loci were associated with kidney- or electrolyte-related traits; these may affect crystallization. Weighted genetic risk score analysis indicated that the highest risk group (top 20%) showed an odds ratio of 1.71 (95% confidence interval, 1.42 to 2.06) - 2.13 (95% confidence interval, 2.00 to 2.27) compared with the reference group (bottom 20%). CONCLUSIONS Our findings provide evidence that host genetic factors related to regulation of metabolic and crystallization pathways contribute to the development of urolithiasis.
Collapse
Affiliation(s)
| | - Yoichiro Kamatani
- Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Chikashi Terao
- Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Masayuki Usami
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Atsushi Takahashi
- Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.,Department of Genomic Medicine, Research Institute, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yukihide Momozawa
- Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Kichiya Suzuki
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Soichi Ogishima
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Atsushi Shimizu
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Mamoru Satoh
- Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Iwate, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Epidemiology and
| | - Haruo Mikami
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan.,Department of Oral Epidemiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | | | | | | | - Shoichiro Tsugane
- Center for Public Health Sciences, National Cancer Center, Tokyo, Japan; and
| | - Kenjiro Kohri
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Alan S L Yu
- Division of Nephrology and Hypertension and.,The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | - Michiaki Kubo
- Division of Genomic Medicine, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan;
| |
Collapse
|
133
|
Nakatochi M, Kanai M, Nakayama A, Hishida A, Kawamura Y, Ichihara S, Akiyama M, Ikezaki H, Furusyo N, Shimizu S, Yamamoto K, Hirata M, Okada R, Kawai S, Kawaguchi M, Nishida Y, Shimanoe C, Ibusuki R, Takezaki T, Nakajima M, Takao M, Ozaki E, Matsui D, Nishiyama T, Suzuki S, Takashima N, Kita Y, Endoh K, Kuriki K, Uemura H, Arisawa K, Oze I, Matsuo K, Nakamura Y, Mikami H, Tamura T, Nakashima H, Nakamura T, Kato N, Matsuda K, Murakami Y, Matsubara T, Naito M, Kubo M, Kamatani Y, Shinomiya N, Yokota M, Wakai K, Okada Y, Matsuo H. Genome-wide meta-analysis identifies multiple novel loci associated with serum uric acid levels in Japanese individuals. Commun Biol 2019; 2:115. [PMID: 30993211 PMCID: PMC6453927 DOI: 10.1038/s42003-019-0339-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/22/2019] [Indexed: 01/05/2023] Open
Abstract
Gout is a common arthritis caused by elevated serum uric acid (SUA) levels. Here we investigated loci influencing SUA in a genome-wide meta-analysis with 121,745 Japanese subjects. We identified 8948 variants at 36 genomic loci (P<5 × 10-8) including eight novel loci. Of these, missense variants of SESN2 and PNPLA3 were predicted to be damaging to the function of these proteins; another five loci-TMEM18, TM4SF4, MXD3-LMAN2, PSORS1C1-PSORS1C2, and HNF4A-are related to cell metabolism, proliferation, or oxidative stress; and the remaining locus, LINC01578, is unknown. We also identified 132 correlated genes whose expression levels are associated with SUA-increasing alleles. These genes are enriched for the UniProt transport term, suggesting the importance of transport-related genes in SUA regulation. Furthermore, trans-ethnic meta-analysis across our own meta-analysis and the Global Urate Genetics Consortium has revealed 15 more novel loci associated with SUA. Our findings provide insight into the pathogenesis, treatment, and prevention of hyperuricemia/gout.
Collapse
Affiliation(s)
- Masahiro Nakatochi
- Data Science Division, Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, 466-8560 Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, 565-0871 Japan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115 USA
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
- Medical Squadron, Air Base Group, Western Aircraft Control and Warning Wing, Japan Air Self-Defense Force, Kasuga, 816-0804 Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
- Department of General Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, 329-0498 Japan
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - Hiroaki Ikezaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, 812-8582 Japan
| | - Norihiro Furusyo
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, 812-8582 Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, 830-0011 Japan
| | - Makoto Hirata
- Laboratory of Genome Technology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Sayo Kawai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Makoto Kawaguchi
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
- Department of Urology, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, 849-8501 Japan
| | - Chisato Shimanoe
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, 849-8501 Japan
| | - Rie Ibusuki
- International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544 Japan
| | - Toshiro Takezaki
- International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544 Japan
| | - Mayuko Nakajima
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Mikiya Takao
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
- Department of Surgery, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Etsuko Ozaki
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, 602-8566 Japan
| | - Daisuke Matsui
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, 602-8566 Japan
| | - Takeshi Nishiyama
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8602 Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8602 Japan
| | - Naoyuki Takashima
- Department of Health Science, Shiga University of Medical Science, Otsu, 520-2192 Japan
| | - Yoshikuni Kita
- Department of Nursing, Tsuruga City College of Nursing, Fukui, 914-8501 Japan
| | - Kaori Endoh
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526 Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, 422-8526 Japan
| | - Hirokazu Uemura
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503 Japan
| | - Kokichi Arisawa
- Department of Preventive Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8503 Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, 464-8681 Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, 464-8681 Japan
- Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Yohko Nakamura
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, 260-8717 Japan
| | - Haruo Mikami
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, 260-8717 Japan
| | - Takashi Tamura
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Hiroshi Nakashima
- Department of Preventive Medicine and Public Health, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Takahiro Nakamura
- Laboratory for Mathematics, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, 162-8655 Japan
| | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639 Japan
| | - Tatsuaki Matsubara
- Department of Internal Medicine, School of Dentistry, Aichi Gakuin University, Nagoya, 464-8651 Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
- Department of Oral Epidemiology, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, 734-8553 Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8507 Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
| | - Mitsuhiro Yokota
- Department of Genome Science, School of Dentistry, Aichi Gakuin University, Nagoya, 464-8651 Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, 466-8550 Japan
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, 565-0871 Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871 Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, 359-8513 Japan
| |
Collapse
|
134
|
Association between SLC2A9 Genetic Variants and Risk of Hyperuricemia in a Uygur Population. Curr Med Sci 2019; 39:243-249. [DOI: 10.1007/s11596-019-2026-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/16/2019] [Indexed: 10/27/2022]
|
135
|
Krajcoviechova A, Marois-Blanchet FC, Troyanov S, Harvey F, Dumas P, Tremblay J, Cifkova R, Awadalla P, Madore F, Hamet P. Uromodulin in a Pathway Between Decreased Renal Urate Excretion and Albuminuria. Am J Hypertens 2019; 32:384-392. [PMID: 30551216 DOI: 10.1093/ajh/hpy190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 11/25/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The mechanism explaining the inverse association between renal urate and albumin excretion remains unclear. First, we evaluated the impact of candidate variants in the main urate transporter genes (i.e., SLC2A9, SLC22A12, ABCG2) on the association between fractional excretion of uric acid (FEUA) and urinary albumin/creatinine ratio (uACR). Second, we examined uromodulin and sodium excretion as mediators of the association between FEUA and uACR. METHODS We performed cross-sectional analysis of 737 French Canadians from the CARTaGENE cohort, a random sample of the Quebec population aged 40-69 years (a total of 20,004 individuals). Individuals with available genotyping and urinary data were obtained from a sub-study including gender-matched pairs with high and low Framingham Risk Score and vascular rigidity index. We further excluded individuals with an estimated glomerular filtration rate <60 ml/min/1.73 m2, glycosuria, and use of confounding medication. A spot urine sample was analyzed. Genotyping was performed using the Illumina Omni2.5-8 BeadChips. Genetic variants were analyzed using an additive model. RESULTS Final analyses included 593 individuals (45.5% of men; mean age 54.3 ± 8.6). We observed an antagonistic interaction between rs13129697 variant of the SLC2A9 gene and FEUA tertiles on uACR (P = 0.002). Using the mediation analysis, uromodulin explained 32%, fractional excretion of sodium (FENa) 44%, and uromodulin together with FENa explained 70% of the inverse relationship between FEUA and uACR. Bootstrapping process confirmed the role of both mediators. CONCLUSIONS Our data suggest that the association of albuminuria with decreased renal urate excretion may be modified by the transporter SLC2A9, and mediated by uromodulin and sodium handling.
Collapse
Affiliation(s)
- Alena Krajcoviechova
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
- Centre de recherche du CHUM, Montréal, Quebec, Canada
| | | | - Stephan Troyanov
- Division of Nephrology, Department of Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Quebec, Canada
| | | | - Pierre Dumas
- Centre de recherche du CHUM, Montréal, Quebec, Canada
| | | | - Renata Cifkova
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
| | - Philip Awadalla
- Centre hospitalier universitaire Sainte-Justine, CHU Sainte-Justine, Montréal, Quebec, Canada
| | - Francois Madore
- Division of Nephrology, Department of Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Quebec, Canada
| | - Pavel Hamet
- Centre de recherche du CHUM, Montréal, Quebec, Canada
| |
Collapse
|
136
|
Roman YM. The Daniel K. Inouye College of Pharmacy Scripts: Perspectives on the Epidemiology of Gout and Hyperuricemia. HAWAI'I JOURNAL OF MEDICINE & PUBLIC HEALTH : A JOURNAL OF ASIA PACIFIC MEDICINE & PUBLIC HEALTH 2019; 78:71-76. [PMID: 30766768 PMCID: PMC6369891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gout is the most common inflammatory arthritic condition affecting more men than women. Hyperuricemia and the deposition of urate crystals into the joints are the hallmarks of gout. The prevalence of gout and hyperuricemia is rising in the United States and world-wide possibly due to the aging population, comorbidities, and other lifestyle factors. Gout and serum uric acid (SUA) levels are highly heritable, underscoring the role of genetics on disease risk and possibly the racial disparities in gout prevalence. However, high consumption of high fructose corn syrup, alcohol, select dietary lifestyles, and use of diuretics are associated with higher SUA levels and increased risk for developing gout. Adopting healthy diet and lifestyle modifications can lower SUA levels. Nonetheless, diet-based approaches for the management of gout should remain a secondary approach to urate lowering therapy.
Collapse
Affiliation(s)
- Youssef M Roman
- Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, HI
| |
Collapse
|
137
|
Köttgen A, Raffler J, Sekula P, Kastenmüller G. Genome-Wide Association Studies of Metabolite Concentrations (mGWAS): Relevance for Nephrology. Semin Nephrol 2019; 38:151-174. [PMID: 29602398 DOI: 10.1016/j.semnephrol.2018.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolites are small molecules that are intermediates or products of metabolism, many of which are freely filtered by the kidneys. In addition, the kidneys have a central role in metabolite anabolism and catabolism, as well as in active metabolite reabsorption and/or secretion during tubular passage. This review article illustrates how the coupling of genomics and metabolomics in genome-wide association analyses of metabolites can be used to illuminate mechanisms underlying human metabolism, with a special focus on insights relevant to nephrology. First, genetic susceptibility loci for reduced kidney function and chronic kidney disease (CKD) were reviewed systematically for their associations with metabolite concentrations in metabolomics studies of blood and urine. Second, kidney function and CKD-associated metabolites reported from observational studies were interrogated for metabolite-associated genetic variants to generate and discuss complementary insights. Finally, insights originating from the simultaneous study of both blood and urine or by modeling intermetabolite relationships are summarized. We also discuss methodologic questions related to the study of metabolite concentrations in urine as well as among CKD patients. In summary, genome-wide association analyses of metabolites using metabolite concentrations quantified from blood and/or urine are a promising avenue of research to illuminate physiological and pathophysiological functions of the kidney.
Collapse
Affiliation(s)
- Anna Köttgen
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
138
|
Narang RK, Topless R, Cadzow M, Gamble G, Stamp LK, Merriman TR, Dalbeth N. Interactions between serum urate-associated genetic variants and sex on gout risk: analysis of the UK Biobank. Arthritis Res Ther 2019; 21:13. [PMID: 30626429 PMCID: PMC6327586 DOI: 10.1186/s13075-018-1787-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/04/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Sex-specific differences in the effect of genetic variants on serum urate levels have been described. The aim of this study was to systematically examine whether serum urate-associated genetic variants differ in their influence on gout risk in men and women. METHODS This research was conducted using the UK Biobank Resource. Thirty single nucleotide polymorphisms (SNPs) associated with serum urate were tested for their association with gout in men and women of European ancestry, aged 40-69 years. Gene-sex interactions for gout risk were analysed using an interaction analysis in logistic regression models. RESULTS Gout was present in 6768 (4.1%) men and 574 (0.3%) women, with an odds ratio (95% confidence interval) for men 13.42 (12.32-14.62) compared with women. In men, experiment-wide association with gout was observed for 21 of the 30 serum urate-associated SNPs tested, and in women for three of the 30 SNPs. Evidence for gene-sex interaction was observed for ABCG2 (rs2231142) and PDZK1 (rs1471633), with the interaction in ABCG2 driven by an amplified effect in men and in PDZK1 by an absence of effect in women. Similar findings were observed in a sensitivity analysis which excluded pre-menopausal women. For the other SNPs tested, no significant gene-sex interactions were observed. CONCLUSIONS In a large population of European ancestry, ABCG2 and PDZK1 gene-sex interactions exist for gout risk, with the serum urate-raising alleles exerting a greater influence on gout risk in men than in women. In contrast, other serum urate-associated genetic variants do not demonstrate significant gene-sex interactions for gout risk.
Collapse
Affiliation(s)
- Ravi K Narang
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Ruth Topless
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9012, New Zealand
| | - Murray Cadzow
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9012, New Zealand
| | - Greg Gamble
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, 2 Riccarton Avenue, Christchurch, 8140, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, 710 Cumberland Street, Dunedin, 9012, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| |
Collapse
|
139
|
Polymorphisms of ABCG2 and SLC22A12 Genes Associated with Gout Risk in Vietnamese Population. ACTA ACUST UNITED AC 2019; 55:medicina55010008. [PMID: 30621105 PMCID: PMC6359270 DOI: 10.3390/medicina55010008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/13/2018] [Accepted: 01/03/2019] [Indexed: 12/20/2022]
Abstract
Background and objective: Gout is a common form of inflammatory arthritis caused by the crystallization of uric acid. Previous studies have demonstrated that the genetic predisposition of gout varies in different ethnic populations. However the association study of genetic variants with gout remains unknown in the Vietnamese population. Our study aimed to assess the relationship between polymorphisms in ABCG2 and SLC22A12 and gout susceptibility in Vietnamese. Materials and methods: Genomic DNA was extracted from blood of a total of 170 patients with gout and 351 healthy controls. We genotyped single nucleotide polymorphisms (SNPs): rs72552713, rs12505410 of the ABCG2 gene and rs11231825, rs7932775 of the SLC22A12 gene using polymerase chain reaction–restriction fragment length polymorphism (PCR–RFLP) and then confirmed 10% of randomly selected subjects by Sanger sequencing. Results: Three SNPs (rs72552713 and rs12505410 and rs11231825) were in accordance with Hardy–Weinberg Equilibrium (HWE) (p > 0.05) while rs7932775 was not (p < 0.05). For rs72552713, CT genotype was significantly different between gout patient and control groups (p < 0.001) and the T allele was associated with an increased risk of gout (OR = 21.19; 95% CI: 3.00–918.96; p < 0.001). Serum uric acid and hyperuricemia differed significantly between CC and CT genotype groups (p = 0.004 and 0.008, respectively). For rs11231825, a protective effect against gout risk was identified in the presence of the C allele when compared with the T allele (OR = 0.712; 95% CI: 0.526–0.964 p = 0.0302). In contrast, no significant difference of allele frequencies between gout patients and controls was detected for rs12505410 (p > 0.05). However, significant differences in serum uric acid and systolic blood pressure were obtained among gout patients. Conclusion: Our results suggest that ABCG2 rs72552713 and SLC22A12 rs11231825 are likely associated with gout in the Vietnamese population in which T allele may be a risk factor for gout susceptibility.
Collapse
|
140
|
Mehmood A, Zhao L, Wang C, Hossen I, Raka RN, Zhang H. Stevia residue extract increases intestinal uric acid excretion via interactions with intestinal urate transporters in hyperuricemic mice. Food Funct 2019; 10:7900-7912. [DOI: 10.1039/c9fo02032b] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The supplementation of STVRE significantly attenuated hyperuricemia and oxidative stress, upregulated ABCG2 and downregulated GLUT9 (protein and mRNA) expression in hyperuricemic mice.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advance Innovation Center for Food Nutrition and Human Health
- Beijing Technology and Business University
- Beijing 100048
- China
- Beijing Engineering and Technology Research Center of Food Additives
| | - Lei Zhao
- Beijing Advance Innovation Center for Food Nutrition and Human Health
- Beijing Technology and Business University
- Beijing 100048
- China
- Beijing Engineering and Technology Research Center of Food Additives
| | - Chengtao Wang
- Beijing Advance Innovation Center for Food Nutrition and Human Health
- Beijing Technology and Business University
- Beijing 100048
- China
- Beijing Engineering and Technology Research Center of Food Additives
| | - Imam Hossen
- Beijing Advance Innovation Center for Food Nutrition and Human Health
- Beijing Technology and Business University
- Beijing 100048
- China
- Beijing Engineering and Technology Research Center of Food Additives
| | - Rifat Nowshin Raka
- Beijing Advance Innovation Center for Food Nutrition and Human Health
- Beijing Technology and Business University
- Beijing 100048
- China
- Beijing Engineering and Technology Research Center of Food Additives
| | - Huimin Zhang
- Beijing Advance Innovation Center for Food Nutrition and Human Health
- Beijing Technology and Business University
- Beijing 100048
- China
- Beijing Engineering and Technology Research Center of Food Additives
| |
Collapse
|
141
|
Tsepilov YA, Sharapov SZ, Zaytseva OO, Krumsiek J, Prehn C, Adamski J, Kastenmüller G, Wang-Sattler R, Strauch K, Gieger C, Aulchenko YS. A network-based conditional genetic association analysis of the human metabolome. Gigascience 2018; 7:5214749. [PMID: 30496450 PMCID: PMC6287100 DOI: 10.1093/gigascience/giy137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 11/06/2018] [Indexed: 12/24/2022] Open
Abstract
Background Genome-wide association studies have identified hundreds of loci that influence a wide variety of complex human traits; however, little is known regarding the biological mechanism of action of these loci. The recent accumulation of functional genomics (“omics”), including metabolomics data, has created new opportunities for studying the functional role of specific changes in the genome. Functional genomic data are characterized by their high dimensionality, the presence of (strong) statistical dependency between traits, and, potentially, complex genetic control. Therefore, the analysis of such data requires specific statistical genetics methods. Results To facilitate our understanding of the genetic control of omics phenotypes, we propose a trait-centered, network-based conditional genetic association (cGAS) approach for identifying the direct effects of genetic variants on omics-based traits. For each trait of interest, we selected from a biological network a set of other traits to be used as covariates in the cGAS. The network can be reconstructed either from biological pathway databases (a mechanistic approach) or directly from the data, using a Gaussian graphical model applied to the metabolome (a data-driven approach). We derived mathematical expressions that allow comparison of the power of univariate analyses with conditional genetic association analyses. We then tested our approach using data from a population-based Cooperative Health Research in the region of Augsburg (KORA) study (n = 1,784 subjects, 1.7 million single-nucleotide polymorphisms) with measured data for 151 metabolites. Conclusions We found that compared to single-trait analysis, performing a genetic association analysis that includes biologically relevant covariates can either gain or lose power, depending on specific pleiotropic scenarios, for which we provide empirical examples. In the context of analyzed metabolomics data, the mechanistic network approach had more power compared to the data-driven approach. Nevertheless, we believe that our analysis shows that neither a prior-knowledge-only approach nor a phenotypic-data-only approach is optimal, and we discuss possibilities for improvement.
Collapse
Affiliation(s)
- Y A Tsepilov
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Lavrentieva Ave. 10, 630090, Russia.,Natural Scince Department, Novosibirsk State University, Novosibirsk, Pirogova Str. 1, 630090, Russia
| | - S Z Sharapov
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Lavrentieva Ave. 10, 630090, Russia.,Natural Scince Department, Novosibirsk State University, Novosibirsk, Pirogova Str. 1, 630090, Russia
| | - O O Zaytseva
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Lavrentieva Ave. 10, 630090, Russia.,Natural Scince Department, Novosibirsk State University, Novosibirsk, Pirogova Str. 1, 630090, Russia
| | - J Krumsiek
- Institute of Computational Biology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany
| | - C Prehn
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany
| | - J Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany.,Institute of Experimental Genetics, Life and Food Science Center Weihenstephan, Technical University of Munich, Freising-Weihenstephan, Arcisstrasse 21, 80333, Germany.,German Center for Diabetes Research, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany
| | - G Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany
| | - R Wang-Sattler
- German Center for Diabetes Research, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany.,Research Unit of Molecular Epidemiology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany.,Institute of Epidemiology II, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany
| | - K Strauch
- Institute of Genetic Epidemiology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany.,Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Butenandstrasse 5, 81377, Germany
| | - C Gieger
- German Center for Diabetes Research, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany.,Research Unit of Molecular Epidemiology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany.,Institute of Epidemiology II, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Ingolstadter Landtrasse 1, 85764, Germany
| | - Y S Aulchenko
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Lavrentieva Ave. 10, 630090, Russia.,Natural Scince Department, Novosibirsk State University, Novosibirsk, Pirogova Str. 1, 630090, Russia.,PolyOmica, 's-Hertogenbosch, Het Vlaggeschip 61, 5237 PA, The Netherlands
| |
Collapse
|
142
|
Schweiger R, Fisher E, Weissbrod O, Rahmani E, Müller-Nurasyid M, Kunze S, Gieger C, Waldenberger M, Rosset S, Halperin E. Detecting heritable phenotypes without a model using fast permutation testing for heritability and set-tests. Nat Commun 2018; 9:4919. [PMID: 30464216 PMCID: PMC6249264 DOI: 10.1038/s41467-018-07276-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 10/26/2018] [Indexed: 01/08/2023] Open
Abstract
Testing for association between a set of genetic markers and a phenotype is a fundamental task in genetic studies. Standard approaches for heritability and set testing strongly rely on parametric models that make specific assumptions regarding phenotypic variability. Here, we show that resulting p-values may be inflated by up to 15 orders of magnitude, in a heritability study of methylation measurements, and in a heritability and expression quantitative trait loci analysis of gene expression profiles. We propose FEATHER, a method for fast permutation-based testing of marker sets and of heritability, which properly controls for false-positive results. FEATHER eliminated 47% of methylation sites found to be heritable by the parametric test, suggesting a substantial inflation of false-positive findings by alternative methods. Our approach can rapidly identify heritable phenotypes out of millions of phenotypes acquired via high-throughput technologies, does not suffer from model misspecification and is highly efficient. Standard approaches for heritability and set testing in statistical genetics rely on parametric models that might not hold in reality and give inflated p-values. Here, the authors develop a fast method for permutation-based testing of marker sets and of heritability that does not suffer from model misspecification.
Collapse
Affiliation(s)
- Regev Schweiger
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 6997801, Israel.
| | - Eyal Fisher
- School of Mathematical Sciences, Department of Statistics, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Omer Weissbrod
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Elior Rahmani
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, 85764, Germany.,Department of Medicine I, Ludwig-Maximilians-Universität, Munich, 80539, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, 80636, Germany
| | - Sonja Kunze
- 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
| | - Christian Gieger
- 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
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, 80636, 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
| | - Saharon Rosset
- School of Mathematical Sciences, Department of Statistics, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Eran Halperin
- Los Angeles, University of California Los Angeles, Los Angeles, 90095, CA, USA.,Department of Anesthesiology and Perioperative Medicine, University of California, Los Angeles, 90095, CA, USA
| |
Collapse
|
143
|
Wang Z, Cui T, Ci X, Zhao F, Sun Y, Li Y, Liu R, Wu W, Yi X, Liu C. The effect of polymorphism of uric acid transporters on uric acid transport. J Nephrol 2018; 32:177-187. [DOI: 10.1007/s40620-018-0546-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/17/2018] [Indexed: 01/09/2023]
|
144
|
Association of common gene variants in glucokinase regulatory protein with cardiorenal disease: A systematic review and meta-analysis. PLoS One 2018; 13:e0206174. [PMID: 30352097 PMCID: PMC6198948 DOI: 10.1371/journal.pone.0206174] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/08/2018] [Indexed: 12/22/2022] Open
Abstract
Background Small-molecules that disrupt the binding between glucokinase and glucokinase regulatory protein (GKRP) in the liver represent a potential new class of glucose-lowering drugs. It will, however, take years before their effects on clinically relevant cardiovascular endpoints are known. The purpose of this study was to estimate the effects of these drugs on cardiorenal outcomes by studying variants in the GKRP gene (GCKR) that mimic glucokinase-GKRP disruptors. Methods The MEDLINE and EMBASE databases were searched for studies reporting on the association between GCKR variants (rs1260326, rs780094, and rs780093) and coronary artery disease (CAD), estimated glomerular filtration rate (eGFR), and chronic kidney disease (CKD). Results In total 5 CAD studies (n = 274,625 individuals), 7 eGFR studies (n = 195,195 individuals), and 4 CKD studies (n = 31,642 cases and n = 408,432 controls) were included. Meta-analysis revealed a significant association between GCKR variants and CAD (OR:1.02 per risk allele, 95%CI:1.00–1.04, p = 0.01). Sensitivity analyses showed that replacement of one large, influential CAD study by two other, partly overlapping studies resulted in similar point estimates, albeit less precise (OR:1.02; 95%CI:0.98–1.06 and OR: 1.02; 95%CI: 0.99–1.04). GCKR was associated with an improved eGFR (+0.49 ml/min, 95%CI:0.10–0.89, p = 0.01) and a trend towards protection from CKD (OR:0.98, 95%CI:0.95–1.01, p = 0.13). Conclusion This study suggests that increased glucokinase-GKRP disruption has beneficial effects on eGFR, but these may be offset by a disadvantageous effect on coronary artery disease risk. Further studies are warranted to elucidate the mechanistic link between hepatic glucose metabolism and eGFR.
Collapse
|
145
|
Parthasarathy P, Vivekanandan S. Urate crystal deposition, prevention and various diagnosis techniques of GOUT arthritis disease: a comprehensive review. Health Inf Sci Syst 2018; 6:19. [PMID: 30333920 DOI: 10.1007/s13755-018-0058-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/25/2018] [Indexed: 12/20/2022] Open
Abstract
Gout is described as difficult in joint sore, uttermost ordinarily in the principal metatarsophalangeal joint, attend from formation of urate monosodium crystallization in a joint space. Analysis might be affirmed by recognizable proof of urate monosodium precious stones in synovial liquid of the influenced joint. There has been expanded enthusiasm for gout in common scholarly and clinical practice settings. The pervasiveness of both hyperuricemia and gout has ascended as most recent decade of time in created nations and in this way weight of gout as expanded. The relationship of hyperuricemia and gout with cardio results for chance of added advantages in mediation on hyperuricemia was featured in this audit. Imaging procedures have ended up being helpful for location of urate statement, even before the primary clinical indications, empowering the assessment of the degree of testimony and giving target estimation of precious stone exhaustion amid urate-bringing down treatment. In advancement, the indication defines the pre diagnostic of gout and associated commodities is advised to prevent the inflammation, that image procedures will assess the weight on statement as well reaction to urinary bringing down clinical procedure in chose patients, lastly amongst last key goal on social insurance for clinical evaluation with gout is to totally project urate gem stores. In spite of the fact that the formal determination is defined with arthrocentesis and resulting examination, CT and ultrasound discoveries on addition of evaluation and execution of infection administration. The standard therapy methodology is available for the patients and whose disease is refractory to standard therapy.
Collapse
Affiliation(s)
| | - S Vivekanandan
- School of Electrical Engineering, VIT University, Vellore, Tamilnadu India
| |
Collapse
|
146
|
Lee HA, Park BH, Park EA, Cho SJ, Kim HS, Park H. Long-term effects of the SLC2A9 G844A and SLC22A12 C246T variants on serum uric acid concentrations in children. BMC Pediatr 2018; 18:296. [PMID: 30189835 PMCID: PMC6127956 DOI: 10.1186/s12887-018-1272-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 08/30/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND We evaluated the effects of two single-nucleotide polymorphisms on UA concentrations in the first decade of life using repeated-measures data. METHODS We included all subjects who were followed-up at least once and for whom we had both UA and genotypic data (i.e., 375, 204, 307, and 363 patients aged 3, 5, 7, and 9 years, respectively). All participated in the Ewha Birth and Growth Cohort study. We used a mixed model analysis to estimate the longitudinal association of serum UA concentration due to the rs3825017 (SLC22A12 c. 246C > T) and rs16890979 (SLC2A9 c. 844G > A) genotypes. RESULTS Overall, the tracking coefficient of UA concentrations in children 3 to 9 years of age was 0.31, and was higher in boys than in girls (0.34 vs. 0.29, respectively). Regarding individual variance, serum UA concentrations decreased as age increased (β = - 0.07, p < 0.05), but there were no significant differences by sex. The effects of rs3825017 on UA concentration were significant in boys, but not in girls. Boys with the T allele of rs3825017 had higher concentrations than their counterparts regardless of the time of follow-up. The rs16890979 genotypes were not significantly associated with serum UA concentration in either sex. CONCLUSION This study showed that rs3825017 in the SLC22A12 gene was associated with UA concentration in childhood.
Collapse
Affiliation(s)
- Hye Ah Lee
- Department of Preventive Medicine, College of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul, 158-710, Korea.,Clinical Trial Center, Mokdong Hospital, Ewha Womans University, Seoul, Korea
| | - Bo Hyun Park
- Department of Preventive Medicine, College of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul, 158-710, Korea
| | - Eun Ae Park
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Su Jin Cho
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hae Soon Kim
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hyesook Park
- Department of Preventive Medicine, College of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-ku, Seoul, 158-710, Korea.
| |
Collapse
|
147
|
Jing J, Ekici AB, Sitter T, Eckardt KU, Schaeffner E, Li Y, Kronenberg F, Köttgen A, Schultheiss UT. Genetics of serum urate concentrations and gout in a high-risk population, patients with chronic kidney disease. Sci Rep 2018; 8:13184. [PMID: 30181573 PMCID: PMC6123425 DOI: 10.1038/s41598-018-31282-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 08/14/2018] [Indexed: 12/22/2022] Open
Abstract
We evaluated genetics of hyperuricemia and gout, their interaction with kidney function and medication intake in chronic kidney disease (CKD) patients. Genome-wide association studies (GWAS) of urate and gout were performed in 4941 CKD patients in the German Chronic Kidney Disease (GCKD) study. Effect estimates of 26 known urate-associated population-based single nucleotide polymorphisms (SNPs) were examined. Interactions of urate-associated variants with urate-altering medications and clinical characteristics of gout were evaluated. Genome-wide significant associations with serum urate and gout were identified for known loci at SLC2A9 and ABCG2, but not for novel loci. Effects of the 26 known SNPs were of similar magnitude in CKD patients compared to population-based individuals, except for SNPs at ABCG2 that showed greater effects in CKD. Gene-medication interactions were not significant when accounting for multiple testing. Associations with gout in specific joints were significant for SLC2A9 rs12498742 in wrists and midfoot joints. Known genetic variants in SLC2A9 and ABCG2 were associated with urate and gout in a CKD cohort, with effect sizes for ABCG2 significantly greater in CKD compared to the general population. CKD patients are at high risk of gout due to reduced kidney function, diuretics intake and genetic predisposition, making treatment to target challenging.
Collapse
Affiliation(s)
- Jiaojiao Jing
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Sitter
- Department of Nephrology and Hypertension, Ludwig-Maximilians University, Munich, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité, University-Medicine, Berlin, Germany
| | - Elke Schaeffner
- Institute of Public Health, Charité, University-Medicine, Berlin, Germany
| | - Yong Li
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University, Innsbruck, Austria
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany.
| | - Ulla T Schultheiss
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Renal Division, Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
148
|
Yang X, Xiao Y, Liu K, Jiao X, Lin X, Wang Y, Zhang Q. Prevalence of hyperuricemia among the Chinese population of the southeast coastal region and association with single nucleotide polymorphisms in urate‑anion exchanger genes: SLC22A12, ABCG2 and SLC2A9. Mol Med Rep 2018; 18:3050-3058. [PMID: 30015934 DOI: 10.3892/mmr.2018.9290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/15/2018] [Indexed: 02/05/2023] Open
Abstract
Genome‑wide association studies identified that a series of genes, including solute carrier family (SLC) 2 member 9 (SLC2A9), SLC 22 member 12 (SLC22A12) and ATP‑binding cassette sub‑family G member 2 (ABCG2) polymorphisms were associated with serum uric acid (SUA) levels in the present study. High incidence rates of hyperuricemia were reported in the Chinese population of the southeast coastal region; however, no evidence has confirmed the genetic association with SUA levels in this region. The present study aimed to investigate the association between uric acid levels and hyperuricemia, and genotypes of the Chinese population of the southeast coastal region. In the present study, a total of 1,056 healthy patients attending routine checkups were employed to investigate the incidence of hyperuricemia; 300 subjects were then randomly selected from the 1,056 patients for the identification of genetic polymorphisms of SLC2A9rs11722228, SLC22A12rs893006 and ABCG2rs2231142 via high‑resolution melting. The present study reported that the incidence rate of hyperuricemia was 32.6% (42.5% in males and 22.7% in females, respectively). The prevalence of ABCG2rs2231142 polymorphisms (CC, CA and AA) was 44.4, 44.8 and 11.8%, respectively; SLC2A9rs11722228 polymorphisms (CC, CT and TT) were reported to be 49.3, 40.3 and 10.3%, respectively. Additionally, SLC22A12rs893006 polymorphisms (CC, CT and TT) were determined to be 57.2, 38.7 and 4.1%, respectively. The SUA levels were observed to be statistically different among each investigated genotype of ABCG2rs2231142 (P=0.047). The A allele was significantly associated with an increased risk of hyperuricemia (odds ratio=2.405 and 1.133 for CA and AA, respectively). The present study reported that high incidence rates of hyperuricemia in the Chinese population of the southeast coastal region may be closely associated with the variants of ABCG2rs2231142. Whether polymorphisms of SLC2A9rs11722228 and SLC22A12rs893006 are involved in hyperuricemia require further investigation.
Collapse
Affiliation(s)
- Xinran Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yingxiu Xiao
- Department of Neurology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Kaixi Liu
- Department of Clinical Laboratory, Shantou Central Hospital, Shantou, Guangdong 515041, P.R. China
| | - Xiaoyang Jiao
- Medical College of Shantou University, Shantou, Guangdong 515043, P.R. China
| | - Xiaozhe Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yongni Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Qiaoxin Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
149
|
Mannino GC, Fuoco A, Marini MA, Spiga R, Di Fatta C, Mancuso E, Perticone F, Andreozzi F, Sesti G. The polymorphism rs35767 at IGF1 locus is associated with serum urate levels. Sci Rep 2018; 8:12255. [PMID: 30115944 PMCID: PMC6095867 DOI: 10.1038/s41598-018-29665-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/12/2018] [Indexed: 01/01/2023] Open
Abstract
Previous studies suggested that the IGF-1/IGF-1 receptor signaling pathway may contribute to regulate uric acid levels. To confirm this hypothesis, we assessed the effects of the IGF-1-raising genetic variant rs35767 on urate levels in serum and urine, and we investigated IGF-1 ability to modulate the expression of transporters involved in reabsorption and secretion of uric acid in the kidney. The study population included 2794 adult Whites. 24-hour urinary uric acid concentration was available for 229 subjects. rs35767 polymorphism was screened using TaqMan genotyping assays. HEK293 (human embryonic kidney-293) cell line was treated with IGF-1 (1, 5, 10, 50 nM) for 24-hours, and differences in the expression of urate transporters were evaluated via Western Blot and real time rtPCR. Individuals carrying the IGF-1-raising allele (rs35767 T) exhibited significantly lower levels of serum urate according to both additive and recessive models, after correction for gender, age, BMI, glucose tolerance, glomerular filtration rate, and anti-hypertensive treatment. TT genotype carriers displayed higher uricosuria than C allele carriers did, after adjusting for confounders. Exposure of HEK293 cells to IGF-1 resulted in a dose-dependent increase of uric acid transporters deputed to uric acid excretion (MRP4, NPT1 and BCRP), and reduction of GLUT9 expression, the major mediator of uric acid reabsorption, both at mRNA and protein level. We observed a significant association between the functional polymorphism rs35767 near IGF1 with serum urate concentrations and we provide a mechanistic explanation supporting a causal role for IGF-1 in the regulation of uric acid homeostasis.
Collapse
Affiliation(s)
- Gaia C Mannino
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Anastasia Fuoco
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Maria A Marini
- Department of Systems Medicine, University of Rome-Tor Vergata, Rome, Italy
| | - Rosangela Spiga
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Concetta Di Fatta
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Elettra Mancuso
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy.
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
150
|
Auberson M, Stadelmann S, Stoudmann C, Seuwen K, Koesters R, Thorens B, Bonny O. SLC2A9 (GLUT9) mediates urate reabsorption in the mouse kidney. Pflugers Arch 2018; 470:1739-1751. [PMID: 30105595 PMCID: PMC6224025 DOI: 10.1007/s00424-018-2190-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/17/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
Uric acid (UA) is a metabolite of purine degradation and is involved in gout flairs and kidney stones formation. GLUT9 (SLC2A9) was previously shown to be a urate transporter in vitro. In vivo, humans carrying GLUT9 loss-of-function mutations have familial renal hypouricemia type 2, a condition characterized by hypouricemia, UA renal wasting associated with kidney stones, and an increased propensity to acute renal failure during strenuous exercise. Mice carrying a deletion of GLUT9 in the whole body are hyperuricemic and display a severe nephropathy due to intratubular uric acid precipitation. However, the precise role of GLUT9 in the kidney remains poorly characterized. We developed a mouse model in which GLUT9 was deleted specifically along the whole nephron in a tetracycline-inducible manner (subsequently called kidney-inducible KO or kiKO). The urate/creatinine ratio was increased as early as 4 days after induction of the KO and no GLUT9 protein was visible on kidney extracts. kiKO mice are morphologically identical to their wild-type littermates and had no spontaneous kidney stones. Twenty-four-hour urine collection revealed a major increase of urate urinary excretion rate and of the fractional excretion of urate, with no difference in urate concentration in the plasma. Polyuria was observed, but kiKO mice were still able to concentrate urine after water restriction. KiKO mice displayed lower blood pressure accompanied by an increased heart rate. Overall, these results indicate that GLUT9 is a crucial player in renal handling of urate in vivo and a putative target for uricosuric drugs.
Collapse
Affiliation(s)
- Muriel Auberson
- Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1011, Lausanne, Switzerland
| | - Sophie Stadelmann
- Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1011, Lausanne, Switzerland
| | - Candice Stoudmann
- Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1011, Lausanne, Switzerland
| | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | | | - Bernard Thorens
- Centre for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Olivier Bonny
- Department of Pharmacology and Toxicology, University of Lausanne, 27 rue du Bugnon, 1011, Lausanne, Switzerland. .,Service of Nephrology, Department of Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|