1
|
Wang S, Ojewunmi OO, Kamiza A, Ramsay M, Morris AP, Chikowore T, Fatumo S, Asimit JL. Accounting for heterogeneity due to environmental sources in meta-analysis of genome-wide association studies. Commun Biol 2024; 7:1512. [PMID: 39543362 PMCID: PMC11564974 DOI: 10.1038/s42003-024-07236-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Meta-analysis of genome-wide association studies (GWAS) across diverse populations offers power gains to identify loci associated with complex traits and diseases. Often heterogeneity in effect sizes across populations will be correlated with genetic ancestry and environmental exposures (e.g. lifestyle factors). We present an environment-adjusted meta-regression model (env-MR-MEGA) to detect genetic associations by adjusting for and quantifying environmental and ancestral heterogeneity between populations. In simulations, env-MR-MEGA has similar or greater association power than MR-MEGA, with notable gains when the environmental factor has a greater correlation with the trait than ancestry. In our analysis of low-density lipoprotein cholesterol in ~19,000 individuals across twelve sex-stratified GWAS from Africa, adjusting for sex, BMI, and urban status, we identify additional heterogeneity beyond ancestral effects for seven variants. Env-MR-MEGA provides an approach to account for environmental effects using summary-level data, making it a useful tool for meta-analyses without the need to share individual-level data.
Collapse
Affiliation(s)
- Siru Wang
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK.
| | - Oyesola O Ojewunmi
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Abram Kamiza
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- The African Computational Genomic (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda
- Malawi Epidemiology and Intervention Research Unit, Lilongwe, Malawi
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Tinashe Chikowore
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Segun Fatumo
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- The African Computational Genomic (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
2
|
Pommereau A, Sassone F, Poli A, De Silvestris M, Scarabottolo L, Zuschlag Y, Licher T, Bärenz F. The development of a novel high-throughput membrane potential assay and a solid-supported membrane (SSM)-based electrophysiological assay to study the pharmacological inhibition of GLUT9/SLC2A9 isoforms in a drug discovery program. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024:100193. [PMID: 39522878 DOI: 10.1016/j.slasd.2024.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
GLUT9/SLC2A9 is a urate transporter and takes a fundamental role in the maintenance of normal serum urate levels. GLUT9 is the sole transporter of reabsorbed urate from renal epithelial cells to blood, thus making it an ideal pharmacological target for the development of urate-lowering drugs. None of the three currently available assays for studying GLUT9 pharmacological inhibition can support a high throughput drug discovery screening campaign. In this manuscript we present two novel assay technologies which can be used in a drug discovery screening cascade for GLUT9: a GLUT9 membrane potential assay for primary screening; and a solid-supported membrane (SSM)-based supported electrophysiological assay for secondary screening.
Collapse
Affiliation(s)
- Antje Pommereau
- Sanofi, Integrated Drug Discovery, Frankfurt am Main, Germany
| | | | | | | | | | - Yasmin Zuschlag
- Sanofi, Integrated Drug Discovery, Frankfurt am Main, Germany
| | - Thomas Licher
- Sanofi, Integrated Drug Discovery, Frankfurt am Main, Germany
| | - Felix Bärenz
- Sanofi, Integrated Drug Discovery, Frankfurt am Main, Germany.
| |
Collapse
|
3
|
Lee S, Shin D. A combination of red and processed meat intake and polygenic risk score influences the incidence of hyperuricemia in middle-aged Korean adults. Nutr Res Pract 2024; 18:721-745. [PMID: 39398885 PMCID: PMC11464275 DOI: 10.4162/nrp.2024.18.5.721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/03/2024] [Accepted: 08/22/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND/OBJECTIVES The high consumption of purine-rich meat is associated with hyperuricemia. However, there is limited evidence linking the consumption of red and processed meat to the genetic risk of hyperuricemia. We investigated the relationship between various combinations of red and processed meat consumption and the polygenic risk scores (PRSs) and the incidence of hyperuricemia in middle-aged Koreans. SUBJECTS/METHODS We analyzed the data from 44,053 participants aged ≥40 years sourced from the Health Examinees (HEXA) cohort of the Korean Genome and Epidemiology Study (KoGES). Information regarding red and processed meat intake was obtained using a semiquantitative food frequency questionnaire (SQ-FFQ). We identified 69 independent single-nucleotide polymorphisms (SNPs) at uric acid-related loci using genome-wide association studies (GWASs) and clumping analyses. The individual PRS, which is the weighted sum of the effect size of each allele at the SNP, was calculated. We used multivariable Cox proportional hazards models adjusted for covariates to determine the relationship between red and processed meat intake and the PRS in the incidence of hyperuricemia. RESULTS During an average follow-up period of 5 years, 2,556 patients with hyperuricemia were identified. For both men and women, the group with the highest red and processed meat intake and the highest PRS was positively associated with the development of hyperuricemia when compared with the group with the lowest red and processed meat intake and the lowest PRS (hazard ratio [HR], 2.72; 95% confidence interval [CI], 2.10-3.53; P < 0.0001; HR, 3.28; 95% CI, 2.45-4.40; P < 0.0001). CONCLUSION Individuals at a high genetic risk for uric acid levels should moderate their consumption of red and processed meat to prevent hyperuricemia.
Collapse
Affiliation(s)
- Suyeon Lee
- Department of Food and Nutrition, Inha University, Incheon 22212, Korea
| | - Dayeon Shin
- Department of Food and Nutrition, Inha University, Incheon 22212, Korea
| |
Collapse
|
4
|
Ren S, Chen S, Huang J, Yu R, Wu Y, Peng XE. Association Between Serum Uric Acid Levels and Metabolic-Associated Fatty Liver Disease in Southeast China: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2024; 17:3343-3354. [PMID: 39268333 PMCID: PMC11390830 DOI: 10.2147/dmso.s476045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Objective This study aimed to explore the association between serum uric acid (sUA) levels and metabolic-associated fatty liver disease (MAFLD) in Southeast China. Methods We performed a cross-sectional study of 2605 subjects who underwent physical examination between 2015 and 2017 in Southeast China. To explore the association between sUA levels and the risk of MAFLD, we employed logistic regression, restricted cubic spline (RCS), subgroups and multiplicative interaction analysis. Results Logistic regression analysis showed a positive association between sUA and MAFLD [aOR total population (95% CI)= 1.90 (1.49 ~ 2.42)], [aOR male (95% CI)= 2.01 (1.54 ~ 2.62)], [aOR female (95% CI)= 1.15 (0.62 ~ 2.11)], respectively. The RCS plot presented a significant nonlinear dose-response relationship between sUA levels and MAFLD risk, and the risk of MAFLD increased significantly when sUA> 5.56 mg/dL (P nonlinear< 0.001). Subgroups analysis revealed that the positive association between sUA and MAFLD was consistent across strata of gender, age, BMI, drinking status, smoking status and tea drinking status. Significant associations between sUA and MAFLD were not only found in males but also existed in subjects whose age ≤60, BMI ≥24 kg/m2, drinkers, smokers and tea-drinkers. Adjusted ORs were estimated to be 2.01, 1.95, 2.11, 2.29, 2.64 and 2.20, respectively. Multiplicative interactions were not observed between gender, age, drinking status, smoking status, tea drinking status and sUA (all P interaction> 0.05). Conclusion According to our study, sUA was positively associated with the risk of MAFLD. Additionally, the risk of MAFLD increased significantly when sUA levels exceeded 5.56 mg/dL. Our study may help clarify whether sUA plays a diagnostic role in MAFLD.
Collapse
Affiliation(s)
- Shutong Ren
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Siyu Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, 361104, People's Republic of China
| | - Jingru Huang
- Department of Clinical Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, People's Republic of China
| | - Rong Yu
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Yunli Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Xian-E Peng
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| |
Collapse
|
5
|
Karimian M, Shabani M, Nikzad H. Association of Functional Genetic Variations in Uric Acid Transporters with the Risk of Idiopathic Male Infertility: A Genetic Association Study and Bioinformatic Analysis. Biochem Genet 2024:10.1007/s10528-024-10902-6. [PMID: 39141156 DOI: 10.1007/s10528-024-10902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
Uric acid plays an important role in sustaining and improving sperm morphology, viability, and motility. It is known that SLC2A9 and ABCG2 protein are the main urate transporter and genetic variations in these genes could be associated with the levels of serum uric acid. This study aimed to investigate the association between single-nucleotide polymorphisms (SNPs) SLC2A9-rs16890979, SLC2A9-rs3733591, ABCG2-rs2231142, and ABCG2-rs2231137 with male infertility. Additionally, the correlation of these SNPs with the uric acid level in seminal plasma of infertile men was examined. Subsequently, an in silico analysis was performed. In a case-control study, 193 infertile and 154 healthy controls were recruited. After semen sample collection, the uric acid level of seminal plasma was measured by a commercial kit. After genomic DNA extraction from sperm samples, SNPs genotyping was performed by PCR-RFLP method. Lastly, the effects of SNPs on the SLC2A9 and ABCG2 gene function were evaluated by bioinformatics tools. The genetic association study revealed that there are significant associations between rs16890979, rs3733591, rs2231142, and rs2231137 genetic variations and increased risk of male infertility. Also, these variations were associated with oligozoospermia and teratozoospermia, and sometimes with asthenozoospermia. Also, we found that four studied SNPs could be associated with a decreased level of uric acid of seminal plasma in teratozoospermia and asthenozoospermia. Bioinformatic analysis revealed that the mentioned polymorphisms could affect molecular aspects of SLC2A9 and ABCG2 genes. In this preliminary study, the rs16890979, rs3733591, rs2231142, and rs2231137 genetic variations could be considered as genetic risk factors for male infertility by interfering with the uric acid level of seminal plasma.
Collapse
Affiliation(s)
- Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, 47416-95447, Iran.
| | - Maryam Shabani
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Qotb-e Ravandi Blvd., Kashan, 8715988141, Iran
| | - Hossein Nikzad
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Qotb-e Ravandi Blvd., Kashan, 8715988141, Iran.
| |
Collapse
|
6
|
Pazokitoroudi A, Liu Z, Dahl A, Zaitlen N, Rosset S, Sankararaman S. A scalable and robust variance components method reveals insights into the architecture of gene-environment interactions underlying complex traits. Am J Hum Genet 2024; 111:1462-1480. [PMID: 38866020 PMCID: PMC11267529 DOI: 10.1016/j.ajhg.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024] Open
Abstract
Understanding the contribution of gene-environment interactions (GxE) to complex trait variation can provide insights into disease mechanisms, explain sources of heritability, and improve genetic risk prediction. While large biobanks with genetic and deep phenotypic data hold promise for obtaining novel insights into GxE, our understanding of GxE architecture in complex traits remains limited. We introduce a method to estimate the proportion of trait variance explained by GxE (GxE heritability) and additive genetic effects (additive heritability) across the genome and within specific genomic annotations. We show that our method is accurate in simulations and computationally efficient for biobank-scale datasets. We applied our method to common array SNPs (MAF ≥1%), fifty quantitative traits, and four environmental variables (smoking, sex, age, and statin usage) in unrelated white British individuals in the UK Biobank. We found 68 trait-E pairs with significant genome-wide GxE heritability (p<0.05/200) with a ratio of GxE to additive heritability of ≈6.8% on average. Analyzing ≈8 million imputed SNPs (MAF ≥0.1%), we documented an approximate 28% increase in genome-wide GxE heritability compared to array SNPs. We partitioned GxE heritability across minor allele frequency (MAF) and local linkage disequilibrium (LD) values, revealing that, like additive allelic effects, GxE allelic effects tend to increase with decreasing MAF and LD. Analyzing GxE heritability near genes highly expressed in specific tissues, we find significant brain-specific enrichment for body mass index (BMI) and basal metabolic rate in the context of smoking and adipose-specific enrichment for waist-hip ratio (WHR) in the context of sex.
Collapse
Affiliation(s)
- Ali Pazokitoroudi
- Department of Computer Science, UCLA, Los Angeles, CA, USA; Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Zhengtong Liu
- Department of Computer Science, UCLA, Los Angeles, CA, USA
| | - Andrew Dahl
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Noah Zaitlen
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Computational Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Neurology, UCLA, Los Angeles, CA, USA
| | - Saharon Rosset
- Department of Statistics, Tel-Aviv University, Tel-Aviv, Israel
| | - Sriram Sankararaman
- Department of Computer Science, UCLA, Los Angeles, CA, USA; Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Computational Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Lin B, Paterson AD, Sun L. Better together against genetic heterogeneity: A sex-combined joint main and interaction analysis of 290 quantitative traits in the UK Biobank. PLoS Genet 2024; 20:e1011221. [PMID: 38656964 PMCID: PMC11073786 DOI: 10.1371/journal.pgen.1011221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/06/2024] [Accepted: 03/11/2024] [Indexed: 04/26/2024] Open
Abstract
Genetic effects can be sex-specific, particularly for traits such as testosterone, a sex hormone. While sex-stratified analysis provides easily interpretable sex-specific effect size estimates, the presence of sex-differences in SNP effect implies a SNP×sex interaction. This suggests the usage of the often overlooked joint test, testing for an SNP's main and SNP×sex interaction effects simultaneously. Notably, even without individual-level data, the joint test statistic can be derived from sex-stratified summary statistics through an omnibus meta-analysis. Utilizing the available sex-stratified summary statistics of the UK Biobank, we performed such omnibus meta-analyses for 290 quantitative traits. Results revealed that this approach is robust to genetic effect heterogeneity and can outperform the traditional sex-stratified or sex-combined main effect-only tests. Therefore, we advocate using the omnibus meta-analysis that captures both the main and interaction effects. Subsequent sex-stratified analysis should be conducted for sex-specific effect size estimation and interpretation.
Collapse
Affiliation(s)
- Boxi Lin
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Andrew D. Paterson
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lei Sun
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Statistical Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Wu S, Li C, Li Y, Liu J, Rong C, Pei H, Li X, Zeng X, Mao W. SLC2A9 rs16890979 reduces uric acid absorption by kidney organoids. Front Cell Dev Biol 2024; 11:1268226. [PMID: 38269090 PMCID: PMC10806012 DOI: 10.3389/fcell.2023.1268226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/14/2023] [Indexed: 01/26/2024] Open
Abstract
Introduction: The excretion and absorption of uric acid (UA) by the kidneys helps regulate serum UA levels. GLUT9, encoded by SLC2A9, is mainly expressed in the renal tubules responsible for UA absorption. SLC2A9 polymorphisms are associated with different serum UA levels. However, the lack of proper in vitro models has stalled research on the mechanisms of single nucleotide polymorphisms (SNPs) that affect UA metabolism in human urate transporters. Methods: In this study, we constructed a gene-edited human embryonic stem cells-9 (ESC-H9) derived kidney organoid bearing rs16890979, an SLC2A9 missense mutation with undetermined associations with hyperuricemia or hypouricemia. Kidney organoids derived from ESC-H9 with genetical overexpression (OE) and low expression (shRNA) of SLC2A9 to serve as controls to study the function of SLC2A9. The function of rs16890979 on UA metabolism was evaluated after placing the organoids to urate-containing medium and following histopathological analysis. Results: The kidney organoids with heterozygous or homozygous rs16890979 mutations showed normal SLC2A9 expression levels and histological distribution, phenotypically similar to the wild-type controls. However, reduced absorption of UA by the kidney organoids with rs16890979 mutants was observed. This finding together with the observation that UA absorption is increased in organoids with SLC2A9 overexpression and decreased in those with SLC2A9 knockdown, suggest that GLUT9 is responsible for UA absorption, and the rs16890979 SNP may compromise this functionality. Moreover, epithelial-mesenchymal transition (EMT) was detected in organoids after UA treatment, especially in the kidney organoid carrying GLUT9OE, suggesting the cytobiological mechanism explaining the pathological features in hyperuricosuria-related renal injury. Discussion: This study showing the transitional value of kidney organoid modeling the function of SNPs on UA metabolism. With a defined genetic background and a confirmed UA absorption function should be useful for studies on renal histological, cellular, and molecular mechanisms with this organoid model.
Collapse
Affiliation(s)
- Shouhai Wu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chuang Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou, China
| | - Yizhen Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junyi Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cuiping Rong
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongfei Pei
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xiong Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Zeng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lab of Stem Cell Biology and Innovative Research of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine/Guangdong Academy of Chinese Medicine, Guangzhou, China
- National Institute for Stem Cell Clinical Research, Guangdong Provincial Hospital of Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Mao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
9
|
Vávra J, Pavelcová K, Mašínová J, Hasíková L, Bubeníková E, Urbanová A, Mančíková A, Stibůrková B. Examining the Association of Rare Allelic Variants in Urate Transporters SLC22A11, SLC22A13, and SLC17A1 with Hyperuricemia and Gout. DISEASE MARKERS 2024; 2024:5930566. [PMID: 38222853 PMCID: PMC10787658 DOI: 10.1155/2024/5930566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/28/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024]
Abstract
Genetic variations in urate transporters play a significant role in determining human urate levels and have been implicated in developing hyperuricemia or gout. Polymorphism in the key urate transporters, such as ABCG2, URAT1, or GLUT9 was well-documented in the literature. Therefore in this study, our objective was to determine the frequency and effect of rare nonsynonymous allelic variants of SLC22A11, SLC22A13, and SLC17A1 on urate transport. In a cohort of 150 Czech patients with primary hyperuricemia and gout, we examined all coding regions and exon-intron boundaries of SLC22A11, SLC22A13, and SLC17A1 using PCR amplification and Sanger sequencing. For comparison, we used a control group consisting of 115 normouricemic subjects. To examine the effects of the rare allelic nonsynonymous variants on the expression, intracellular processing, and urate transporter protein function, we performed a functional characterization using the HEK293A cell line, immunoblotting, fluorescent microscopy, and site directed mutagenesis for preparing variants in vitro. Variants p.V202M (rs201209258), p.R343L (rs75933978), and p.P519L (rs144573306) were identified in the SLC22A11 gene (OAT4 transporter); variants p.R16H (rs72542450), and p.R102H (rs113229654) in the SLC22A13 gene (OAT10 transporter); and the p.W75C variant in the SLC17A1 gene (NPT1 transporter). All variants minimally affected protein levels and cytoplasmic/plasma membrane localization. The functional in vitro assay revealed that contrary to the native proteins, variants p.P519L in OAT4 (p ≤ 0.05), p.R16H in OAT10 (p ≤ 0.05), and p.W75C in the NPT1 transporter (p ≤ 0.01) significantly limited urate transport activity. Our findings contribute to a better understanding of (1) the risk of urate transporter-related hyperuricemia/gout and (2) uric acid handling in the kidneys.
Collapse
Affiliation(s)
- Jiří Vávra
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | | | | | | | - Eliška Bubeníková
- Institute of Rheumatology, Prague, Czech Republic
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Aneta Urbanová
- 1st Department of Medicine, Department of Hematology; First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Andrea Mančíková
- Department of Staphylococcal and Food-Borne Bacterial Infections, The National Institute of Public Health, Prague, Czech Republic
| | - Blanka Stibůrková
- Institute of Rheumatology, Prague, Czech Republic
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
10
|
Ma N, Cai S, Sun Y, Chu C. Chinese Sumac ( Rhus chinensis Mill.) Fruits Prevent Hyperuricemia and Uric Acid Nephropathy in Mice Fed a High-Purine Yeast Diet. Nutrients 2024; 16:184. [PMID: 38257077 PMCID: PMC10819650 DOI: 10.3390/nu16020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Hyperuricemia (HUA) is a prevalent chronic disease, characterized by excessive blood uric acid levels, that poses a significant health risk. In this study, the preventive effects and potential mechanisms of ethanol extracts from Chinese sumac (Rhus chinensis Mill.) fruits on HUA and uric acid nephropathy were comprehensively investigated. The results demonstrated a significant reduction in uric acid levels in hyperuricemia mice after treatment with Chinese sumac fruit extract, especially in the high-dose group, where the blood uric acid level decreased by 39.56%. Visual diagrams of the kidneys and hematoxylin and eosin (H&E)-stained sections showed the extract's effectiveness in protecting against kidney damage caused by excessive uric acid. Further investigation into its mechanism revealed that the extract prevents and treats hyperuricemia by decreasing uric acid production, enhancing uric acid excretion, and mitigating the oxidative stress and inflammatory reactions induced by excessive uric acid in the kidneys. Specifically, the extract markedly decreased xanthine oxidase (XOD) levels and expression in the liver, elevated the expression of uric acid transporters ABCG2, and lowered the expression of uric acid reabsorption proteins URAT1 and SLC2A9. Simultaneously, it significantly elevated the levels of endogenous antioxidant enzymes (SOD and GSH) while reducing the level of malondialdehyde (MDA). Furthermore, the expression of uric-acid-related proteins NLRP3, ACS, and Caspase-3 and the levels of IL-1β and IL-6 were significantly reduced. The experimental results confirm that Chinese sumac fruit extract can improve HUA and uric acid nephropathy in mice fed a high-purine yeast diet. This finding establishes a theoretical foundation for developing Chinese sumac fruit as a functional food or medicine for preventing and treating HUA.
Collapse
Affiliation(s)
| | | | | | - Chuanqi Chu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; (N.M.); (S.C.)
| |
Collapse
|
11
|
Ichida K. [Uric Acid Metabolism, Uric Acid Transporters and Dysuricemia]. YAKUGAKU ZASSHI 2024; 144:659-674. [PMID: 38825475 DOI: 10.1248/yakushi.23-00217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Serum urate levels are determined by the balance between uric acid production and uric acid excretion capacity from the kidneys and intestinal tract. Dysuricemia, including hyperuricemia and hypouricemia, develops when the balance shifts towards an increase or a decrease in the uric acid pool. Hyperuricemia is mostly a multifactorial genetic disorder involving several disease susceptibility genes and environmental factors. Hypouricemia, on the other hand, is caused by genetic abnormalities. The main genes involved in dysuricemia are xanthine oxidoreductase, an enzyme that produces uric acid, and the urate transporters urate transporter 1/solute carrier family 22 member 12 (URAT1/SLC22A12), glucose transporter 9/solute carrier family 2 member 9 (GLUT9/SLC2A9) and ATP binding cassette subfamily G member 2 (ABCG2). Deficiency of xanthine oxidoreductase results in xanthinuria, a rare disease with marked hypouricemia. Xanthinuria can be due to a single deficiency of xanthine oxidoreductase or in combination with aldehyde oxidase deficiency as well. The latter is caused by a deficiency in molybdenum cofactor sulfurase, which is responsible for adding sulphur atoms to the molybdenum cofactor required for xanthine oxidoreductase and aldehyde oxidase to exert their action. URAT1/SLC22A12 and GLUT9/SLC2A9 are involved in urate reabsorption and their deficiency leads to renal hypouricemia, a condition that is common in Japanese due to URAT1/SLC22A12 deficiency. On the other hand, ABCG2 is involved in the secretion of urate, and many Japanese have single nucleotide polymorphisms that result in its reduced function, leading to hyperuricemia. In particular, severe dysfunction of ABCG2 leads to hyperuricemia with reduced extrarenal excretion.
Collapse
MESH Headings
- Humans
- Hyperuricemia/etiology
- Hyperuricemia/metabolism
- Hyperuricemia/genetics
- Uric Acid/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Organic Anion Transporters/metabolism
- Organic Anion Transporters/genetics
- Glucose Transport Proteins, Facilitative/metabolism
- Glucose Transport Proteins, Facilitative/genetics
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Xanthine Dehydrogenase/metabolism
- Xanthine Dehydrogenase/genetics
- Xanthine Dehydrogenase/deficiency
- Animals
- Organic Cation Transport Proteins/genetics
- Organic Cation Transport Proteins/metabolism
- Renal Tubular Transport, Inborn Errors/genetics
- Renal Tubular Transport, Inborn Errors/etiology
- Renal Tubular Transport, Inborn Errors/metabolism
- Urinary Calculi/etiology
- Urinary Calculi/metabolism
- Urinary Calculi/genetics
- Metabolism, Inborn Errors
Collapse
Affiliation(s)
- Kimiyoshi Ichida
- Department of Pathophysiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
- Division of Kidney and Hypertension, The Jikei University School of Medicine
| |
Collapse
|
12
|
Miao J, Wu Y, Lu Q. Statistical methods for gene-environment interaction analysis. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL STATISTICS 2024; 16:e1635. [PMID: 38699459 PMCID: PMC11064894 DOI: 10.1002/wics.1635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/12/2023] [Indexed: 05/05/2024]
Abstract
Most human complex phenotypes result from multiple genetic and environmental factors and their interactions. Understanding the mechanisms by which genetic and environmental factors interact offers valuable insights into the genetic architecture of complex traits and holds great potential for advancing precision medicine. The emergence of large population biobanks has led to the development of numerous statistical methods aiming at identifying gene-environment interactions (G × E). In this review, we present state-of-the-art statistical methodologies for G × E analysis. We will survey a spectrum of approaches for single-variant G × E mapping, followed by various techniques for polygenic G × E analysis. We conclude this review with a discussion on the future directions and challenges in G × E research.
Collapse
Affiliation(s)
- Jiacheng Miao
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Yixuan Wu
- University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Department of Statistics, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Center for Demography of Health and Aging, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
13
|
Kim JH, Yang HJ, Park S, Lee HJ, Song YS. Differential Gene Expression in the Penile Cavernosum of Streptozotocin-Induced Diabetic Rats. Int Neurourol J 2023; 27:234-242. [PMID: 38171323 PMCID: PMC10762368 DOI: 10.5213/inj.2346074.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/01/2023] [Indexed: 01/05/2024] Open
Abstract
PURPOSE Men with diabetes mellitus (DM) often present with severe erectile dysfunction (ED). This ED is less responsive to current pharmacological therapies. If we know the upregulated or downregulated genes of diabetic ED, we can inhibit or enhance the expression of such genes through RNA or gene overexpression. METHODS To investigate gene changes associated with ED in type 1 DM, we examined the alterations of gene expression in the cavernosum of streptozotocin-induced diabetic rats. Specifically, we considered 11,636 genes (9,623 upregulated and 2,013 downregulated) to be differentially expressed in the diabetic rat cavernosum group (n=4) compared to the control group (n=4). The analysis of differentially expressed genes using the gene ontology (GO) classification indicated that the following were enriched: downregulated genes such as cell cycle, extracellular matrix, glycosylphosphatidylinositol-anchor biosynthesis and upregulated genes such as calcium signaling, neurotrophin signaling, apoptosis, arginine and proline metabolism, gap junction, transforming growth factor-β signaling, tight junction, vascular smooth muscle contraction, and vascular endothelial growth factor (VEGF) signaling. We examined a more than 2-fold upregulated or downregulated change in expression, using real time polymerase chain reaction. Analysis of differentially expressed genes, using the GO classification, indicated the enrichment. RESULTS Of the 41,105 genes initially considered, statistical filtering of the array analysis showed 9,623 upregulated genes and 2,013 downregulated genes with at least 2-fold changes in expression (P<0.05). With Bonferroni correction, SLC2A9 (solute carrier family 2 member 9), LRRC20 (leucine rick repeat containing 20), PLK1 (polo like kinase 1), and AATK (apoptosis-associated tyrosine kinase) were all 2-fold changed genes. CONCLUSION This study broadens the scope of candidate genes that may be relevant to the pathophysiology of diabetic ED. In particular, their enhancement or inhibition could represent a novel treatment for diabetic ED.
Collapse
Affiliation(s)
- Jae Heon Kim
- Department of Urology, Soonchunhyang University Seoul Hospital, Soonchunhyang University School of Medicine, Seoul, Korea
| | - Hee Jo Yang
- Department of Urology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University School of Medicine, Cheonan, Korea
| | - Suyeon Park
- Department of Biostatistics, Soonchunhyang University School of Medicine, Seoul, Korea
| | - Hong Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea
- Research Institute, e-biogen Inc., Seoul, Korea
| | - Yun Seob Song
- Department of Urology, Soonchunhyang University Seoul Hospital, Soonchunhyang University School of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Fu B, Pazokitoroudi A, Sudarshan M, Liu Z, Subramanian L, Sankararaman S. Fast kernel-based association testing of non-linear genetic effects for biobank-scale data. Nat Commun 2023; 14:4936. [PMID: 37582955 PMCID: PMC10427662 DOI: 10.1038/s41467-023-40346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/18/2023] [Indexed: 08/17/2023] Open
Abstract
Our knowledge of non-linear genetic effects on complex traits remains limited, in part, due to the modest power to detect such effects. While kernel-based tests offer a versatile approach to test for non-linear relationships between sets of genetic variants and traits, current approaches cannot be applied to Biobank-scale datasets containing hundreds of thousands of individuals. We propose, FastKAST, a kernel-based approach that can test for non-linear effects of a set of variants on a quantitative trait. FastKAST provides calibrated hypothesis tests while enabling analysis of Biobank-scale datasets with hundreds of thousands of unrelated individuals from a homogeneous population. We apply FastKAST to 53 quantitative traits measured across ≈ 300 K unrelated white British individuals in the UK Biobank to detect sets of variants with non-linear effects at genome-wide significance.
Collapse
Affiliation(s)
- Boyang Fu
- Department of Computer Science, UCLA, Los Angeles, CA, USA.
| | | | - Mukund Sudarshan
- Department of Computer Science, Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
| | - Zhengtong Liu
- Department of Computer Science, UCLA, Los Angeles, CA, USA
| | - Lakshminarayanan Subramanian
- Department of Computer Science, Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Sriram Sankararaman
- Department of Computer Science, UCLA, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
- Department of Computational Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Toyoda Y, Miyata H, Shigesawa R, Matsuo H, Suzuki H, Takada T. SVCT2/SLC23A2 is a sodium-dependent urate transporter: functional properties and practical application. J Biol Chem 2023; 299:104976. [PMID: 37390985 PMCID: PMC10374969 DOI: 10.1016/j.jbc.2023.104976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/10/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023] Open
Abstract
Urate transporters play a pivotal role in urate handling in the human body, but the urate transporters identified to date do not account for all known molecular processes of urate handling, suggesting the presence of latent machineries. We recently showed that a urate transporter SLC2A12 is also a physiologically important exporter of ascorbate (the main form of vitamin C in the body) that would cooperate with an ascorbate importer, sodium-dependent vitamin C transporter 2 (SVCT2). Based on the dual functions of SLC2A12 and cooperativity between SLC2A12 and SVCT2, we hypothesized that SVCT2 might be able to transport urate. To test this proposal, we conducted cell-based analyses using SVCT2-expressing mammalian cells. The results demonstrated that SVCT2 is a novel urate transporter. Vitamin C inhibited SVCT2-mediated urate transport with a half-maximal inhibitory concentration of 36.59 μM, suggesting that the urate transport activity may be sensitive to physiological ascorbate levels in blood. Similar results were obtained for mouse Svct2. Further, using SVCT2 as a sodium-dependent urate importer, we established a cell-based urate efflux assay that will be useful for identification of other novel urate exporters as well as functional characterization of nonsynonymous variants of already-identified urate exporters including ATP-binding cassette transporter G2. While more studies will be needed to elucidate the physiological impact of SVCT2-mediated urate transport, our findings deepen understanding of urate transport machineries.
Collapse
Affiliation(s)
- Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Miyata
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Ryuichiro Shigesawa
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
16
|
Wang C, Wang J, Wan R, Yuan T, Yang L, Zhang D, Li X, Wang M, Liu H, Lei Y, Wei H, Li J, Liu M, Hua Y, Sun L, Zhang L. Relationship between baseline and changed serum uric acid and the incidence of type 2 diabetes mellitus: a national cohort study. Front Public Health 2023; 11:1170792. [PMID: 37483942 PMCID: PMC10357007 DOI: 10.3389/fpubh.2023.1170792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
Objective To explore the correlation between baseline serum uric acid (SUA) and SUA changes with the incidence of type 2 diabetes mellitus (T2DM) among middle-aged and older individuals. Method Binary logistic regression was used to calculate the odds ratio (ORs) and 95% confidence intervals (CIs) of the effects of baseline and changes in SUA on the incidence of T2DM. Stratified analysis was conducted based on sex, and the SUA levels were classified into four quartiles to assess the effect of baseline and relative changes in SUA on the incidence of T2DM. Furthermore, interaction analysis was performed between body mass index (BMI) and SUA, age and SUA, and sex and SUA. Results In the cohort study, the highest quartiles of SUA were significantly correlated with an increased incidence of T2DM among females in model 1 [OR = 2.231 (1.631, 3.050)], model 2 [OR = 2.090 (1.523, 2.867)], model 3 [OR = 2.075 (1.511, 2.849)], and model 4 [OR = 1.707 (1.234, 2.362)]. The highest quartiles of SUA had a statistically significant effect on the incidence of T2DM among all participants in model 1 [OR = 1.601 (1.277, 2.008)], model 2 [OR = 1.519 (1.204, 1.915)], model 3 [OR = 1.597 (1.257, 2.027)], and model 4 [OR = 1.380 (1.083, 1.760)]. Regarding the relative change of SUA, the highest quantiles of SUA were significantly correlated with an increased incidence of T2DM among females in model 1 [OR = 1.409 (1.050, 1.890)], model 2 [OR = 1.433 (1.067, 1.926)], and model 3 [OR = 1.420 (1.056, 1.910)], and there was a statistically significant correlation with incident T2DM among all participants in model 4 [OR = 1.346 (1.079, 1.680)] after adjusting for all covariates. However, there was no significant correlation between baseline, relative, and absolute changes in SUA and the incidence of T2DM among males. The interaction analysis demonstrated that sex, BMI, and the relative changes in SUA had a combined effect on the incidence of T2DM, while age and the changes in SUA had a joint effect on the incidence of T2DM only in females. Conclusion There was a positive association between SUA and the incidence of T2DM for all participants. However, significant sex differences in incidence were observed only in women, not men.
Collapse
Affiliation(s)
- Congzhi Wang
- Department of Internal Medicine Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Jiazhi Wang
- Sports Institute, Chi Zhou College, Education Park, Chi Zhou, China
| | - Rui Wan
- Business School, Yunnan University of Finance and Economics, Kunming, China
| | - Ting Yuan
- Obstetrics and Gynecology Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Liu Yang
- Department of Internal Medicine Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Dongmei Zhang
- Department of Pediatric Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Xiaoping Li
- Department of Emergency and Critical Care Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Min Wang
- Department of Pharmacy, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Haiyang Liu
- Student Health Center, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Yunxiao Lei
- Obstetrics and Gynecology Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Huanhuan Wei
- Obstetrics and Gynecology Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Jing Li
- Department of Surgical Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Mingming Liu
- Department of Surgical Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Ying Hua
- Rehabilitation Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Lu Sun
- Department of Emergency and Critical Care Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| | - Lin Zhang
- Department of Internal Medicine Nursing, School of Nursing, Wannan Medical College, Higher Education Park, Wuhu, China
| |
Collapse
|
17
|
Xie Y, Huang K, Zhang X, Wu Z, Wu Y, Chu J, Kong W, Qian G. Association of serum uric acid-to-high-density lipoprotein cholesterol ratio with non-alcoholic fatty liver disease in American adults: a population-based analysis. Front Med (Lausanne) 2023; 10:1164096. [PMID: 37256087 PMCID: PMC10225665 DOI: 10.3389/fmed.2023.1164096] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/14/2023] [Indexed: 06/01/2023] Open
Abstract
Objective Non-invasive disease indicators are currently limited and need further research due to the increased non-alcoholic fatty liver disease (NAFLD) prevalence worldwide. The serum uric acid-to-high-density lipoprotein cholesterol ratio (UHR) has been recognized as a novel inflammatory and metabolic marker. Herein, we explored the correlation between UHR and the risk of NAFLD in-depth. Methods A total of 3,766 participants were included in our survey, and the National Health and Nutrition Examination Survey (NHANES) 2017-2018 cycle provided the cross-sectional study population. Weighted multivariable logistic regression and multivariate linear regression analyses were performed to assess the association between the UHR and the odds of NAFLD and liver steatosis and fibrosis severity, respectively. Moreover, we explored the non-linear relationship between the UHR and NAFLD by the generalized additive model. Results NAFLD probabilities were statistically demonstrated to be positively correlated with the UHR (OR = 1.331 per SD increase, 95% CI: 1.100, 1.611). The positive connection of the UHR with NAFLD risk persisted significantly in female subjects but not in male subjects in subgroup analyses stratified by gender. The non-linear relationship analysis demonstrated that a UHR between ~20 and 30% suggested a saturation effect of NAFLD risk. Furthermore, a dramatically positive correlation was found between the UHR and hepatic steatosis severity but not fibrosis. Finally, the receiver operating characteristic analysis suggested that UHR had a better predictive value for NAFLD than either serum uric acid (sUA) or high-density lipoprotein cholesterol (HDL) alone [UHR (area under curve): 0.6910; 95% CI: 0.6737-0.7083; P < 0.0001]. Conclusion Our investigation revealed that the elevated UHR level was independently related to an increased NAFLD risk and the severity of liver steatosis in American individuals. The correlation differed according to sex. This non-invasive indicator may enhance the capacity to predict the onset of NAFLD and may uncover alternative therapeutic interventional targets.
Collapse
Affiliation(s)
- Yilian Xie
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo, Zhejiang, China
- Department of Hepatology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Kai Huang
- Department of General Medicine, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Xiangyu Zhang
- Department of Nephrology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Zhouxiao Wu
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo, Zhejiang, China
- Department of Hepatology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Yiyi Wu
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo, Zhejiang, China
- Department of Hepatology, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Jinguo Chu
- Department of Hepatology, Ningbo First Hospital, Ningbo, Zhejiang, China
- Department of General Medicine, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Weiliang Kong
- Department of Respiratory and Critical Care Medicine, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Guoqing Qian
- Department of Infectious Diseases, Ningbo First Hospital, Ningbo, Zhejiang, China
- Department of Hepatology, Ningbo First Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
18
|
Halengbieke A, Zhang S, Tong C, Ni XT, Han YM, Zheng DQ, Tao LX, Guo XH, Li Q, Yang XH. Causal relationship between serum uric acid and abnormal blood pressure based on the panel model study: A 5-year cohort study. Nutr Metab Cardiovasc Dis 2023; 33:500-506. [PMID: 36646600 DOI: 10.1016/j.numecd.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/07/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND AIMS To investigate the relationship between elevated serum uric acid (SUA) levels and blood pressure (BP). METHODS AND RESULTS Based on the Beijing Health Management Cohort, 5276 health examination people were enrolled. Cross-lagged model was used to explore the relationship between SUA levels and blood pressure. The results showed: (1) increased SUA and increased systolic blood pressure (SBP): ① The path coefficients from baseline SUA to follow-up SBP were statistically significant in both the general population (β = 0.034, P < 0.05) and men (β = 0.048, P < 0.05). The path coefficients from baseline SBP to follow-up SUA were not statistically significant in either the general population (β = 0.010, P > 0.05) or men (β = 0.011, P > 0.05). ② The path coefficients from baseline SUA to follow-up SBP and from baseline SBP to follow-up SUA were not statistically significant in women with BMI ≥ 25 kg/m2 and BMI < 25 kg/m2. (2) Increased SUA and diastolic blood pressure (DBP): ① There was no statistical significance between the path coefficients from baseline DBP to follow-up SUA and the path coefficients from baseline SUA to follow-up DBP. ② In men and women, BMI ≥ 25 kg/m2 and BMI < 25 kg/m2, the path coefficients from baseline DBP to follow-up SUA and from baseline SUA to follow-up DBP were not statistically significant. CONCLUSIONS SUA can increase blood pressure in the general male population; no reverse time sequence relationship was found. The temporal relationships between SUA levels and SBP abnormalities were different in the sex and BMI subgroups. No bidirectional causal temporal relationship was found between SUA elevation and DBP abnormality.
Collapse
Affiliation(s)
- Aheyeerke Halengbieke
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Shan Zhang
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Chao Tong
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Xue Tong Ni
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Yu Mei Han
- Beijing Physical Examination Center, China.
| | - De Qiang Zheng
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Li Xin Tao
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Xiu Hua Guo
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Qiang Li
- Beijing Physical Examination Center, China.
| | - Xing Hua Yang
- School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| |
Collapse
|
19
|
Mandal AK, Leask MP, Sumpter NA, Choi HK, Merriman TR, Mount DB. Genetic and Physiological Effects of Insulin-Like Growth Factor-1 (IGF-1) on Human Urate Homeostasis. J Am Soc Nephrol 2023; 34:451-466. [PMID: 36735516 PMCID: PMC10103387 DOI: 10.1681/asn.0000000000000054] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/25/2022] [Indexed: 01/22/2023] Open
Abstract
SIGNIFICANCE STATEMENT Hyperinsulinemia induces hyperuricemia by activating net renal urate reabsorption in the renal proximal tubule. The basolateral reabsorptive urate transporter GLUT9a appears to be the dominant target for insulin. By contrast, IGF-1 infusion reduces serum urate (SU), through mechanisms unknown. Genetic variants of IGF1R associated with reduced SU have increased IGF-1R expression and interact with genes encoding the GLUT9 and ABCG2 urate transporters, in a sex-specific fashion, which controls the SU level. Activation of IGF-1/IGF-1R signaling in Xenopus oocytes modestly activates GLUT9a and inhibits insulin's stimulatory effect on the transporter, which also activates multiple secretory urate transporters-ABCG2, ABCC4, OAT1, and OAT3. The results collectively suggest that IGF-1 reduces SU by activating secretory urate transporters and inhibiting insulin's action on GLUT9a. BACKGROUND Metabolic syndrome and hyperinsulinemia are associated with hyperuricemia. Insulin infusion in healthy volunteers elevates serum urate (SU) by activating net urate reabsorption in the renal proximal tubule, whereas IGF-1 infusion reduces SU by mechanisms unknown. Variation within the IGF1R gene also affects SU levels. METHODS Colocalization analyses of a SU genome-wide association studies signal at IGF1R and expression quantitative trait loci signals in cis using COLOC2, RT-PCR, Western blotting, and urate transport assays in transfected HEK 293T cells and in Xenopus laevis oocytes. RESULTS Genetic association at IGF1R with SU is stronger in women and is mediated by control of IGF1R expression. Inheritance of the urate-lowering homozygous genotype at the SLC2A9 locus is associated with a differential effect of IGF1R genotype between men and women. IGF-1, through IGF-1R, stimulated urate uptake in human renal proximal tubule epithelial cells and transfected HEK 293T cells, through activation of IRS1, PI3/Akt, MEK/ERK, and p38 MAPK; urate uptake was inhibited in the presence of uricosuric drugs, specific inhibitors of protein tyrosine kinase, PI3 kinase (PI3K), ERK, and p38 MAPK. In X. laevis oocytes expressing ten individual urate transporters, IGF-1 through endogenous IGF-1R stimulated urate transport mediated by GLUT9, OAT1, OAT3, ABCG2, and ABCC4 and inhibited insulin's stimulatory action on GLUT9a and OAT3. IGF-1 significantly activated Akt and ERK. Specific inhibitors of PI3K, ERK, and PKC significantly affected IGF-1 stimulation of urate transport in oocytes. CONCLUSIONS The combined results of infusion, genetics, and transport experiments suggest that IGF-1 reduces SU by activating urate secretory transporters and inhibiting insulin's action.
Collapse
Affiliation(s)
- Asim K. Mandal
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Megan P. Leask
- Biochemistry Department, University of Otago, Dunedin, South Island, New Zealand
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, Alabama
| | - Nicholas A. Sumpter
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, Alabama
| | - Hyon K. Choi
- Division of Rheumatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tony R. Merriman
- Biochemistry Department, University of Otago, Dunedin, South Island, New Zealand
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, Alabama
| | - David B. Mount
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Renal Division, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Toyoda Y, Cho SK, Tasic V, Pavelcová K, Bohatá J, Suzuki H, David VA, Yoon J, Pallaiova A, Šaligová J, Nousome D, Cachau R, Winkler CA, Takada T, Stibůrková B. Identification of a dysfunctional exon-skipping splice variant in GLUT9/ SLC2A9 causal for renal hypouricemia type 2. Front Genet 2023; 13:1048330. [PMID: 36733941 PMCID: PMC9887137 DOI: 10.3389/fgene.2022.1048330] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
Renal hypouricemia (RHUC) is a pathological condition characterized by extremely low serum urate and overexcretion of urate in the kidney; this inheritable disorder is classified into type 1 and type 2 based on causative genes encoding physiologically-important urate transporters, URAT1 and GLUT9, respectively; however, research on RHUC type 2 is still behind type 1. We herein describe a typical familial case of RHUC type 2 found in a Slovak family with severe hypouricemia and hyperuricosuria. Via clinico-genetic analyses including whole exome sequencing and in vitro functional assays, we identified an intronic GLUT9 variant, c.1419+1G>A, as the causal mutation that could lead the expression of p.Gly431GlufsTer28, a functionally-null variant resulting from exon 11 skipping. The causal relationship was also confirmed in another unrelated Macedonian family with mild hypouricemia. Accordingly, non-coding regions should be also kept in mind during genetic diagnosis for hypouricemia. Our findings provide a better pathogenic understanding of RHUC and pathophysiological importance of GLUT9.
Collapse
Affiliation(s)
- Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Sung Kweon Cho
- Molecular Genetics Epidemiology Section, Basic Research Laboratory, National Cancer Institute and Frederick National Laboratory for Cancer Research, Frederick, MD, United States,Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea
| | - Velibor Tasic
- Faculty of Medicine, University Ss. Cyril and Methodius, Skopje, North Macedonia
| | | | | | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Victor A. David
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Jaeho Yoon
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | | | - Jana Šaligová
- Metabolic Clinic, Children’s Faculty Hospital, Košice, Slovakia
| | - Darryl Nousome
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Raul Cachau
- Integrated Data Science Section, Research Technologies Branch, National Institute of Allergies and Infectious Diseases, Bethesda, MD, United States
| | - Cheryl A. Winkler
- Molecular Genetics Epidemiology Section, Basic Research Laboratory, National Cancer Institute and Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Blanka Stibůrková
- Institute of Rheumatology, Prague, Czechia,Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czechia,Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia,*Correspondence: Blanka Stibůrková,
| |
Collapse
|
21
|
Chen N, Hu LK, Sun Y, Dong J, Chu X, Lu YK, Liu YH, Ma LL, Yan YX. Associations of waist-to-height ratio with the incidence of type 2 diabetes and mediation analysis: Two independent cohort studies. Obes Res Clin Pract 2023; 17:9-15. [PMID: 36586764 DOI: 10.1016/j.orcp.2022.12.005] [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] [Received: 07/21/2022] [Revised: 12/11/2022] [Accepted: 12/18/2022] [Indexed: 12/31/2022]
Abstract
AIM To assess the relationship between waist-to-height ratio (WHtR) and the incidence of type 2 diabetes (T2D)/impaired fasting glucose (IFG) and to explore to what extent these associations are mediated by blood pressure, lipids and other indicators related to liver and kidney metabolism. MATERIALS AND METHODS This study was based on a functional community cohort included 6109 participants which were divided into two sub-cohorts. One sub-cohort included participants with normal fasting glucose (n = 5563), another included IFG individuals at baseline (n = 546). Cox regression models were used to evaluate the relationships of WHtR with T2D/IFG. Four-year time-dependent receiver operating characteristic (ROC) curve and area under curve (AUC) were calculated to estimate the discriminatory power of WhtR and other anthropometric indices on T2D. Mediation analysis was performed to estimate which risk factors mediate the association between WHtR and T2D. RESULTS Significant positive associations were found between WHtR and the incidence of T2D/IFG in both sub-cohort. WhtR was a useful predictor of T2D (P < 0.05). Mediation analysis showed that HOMA-IR (0.45 %), SBP (5.10 %), triglycerides (11.02 %), creatinine (9.36 %) and combined kidney indicators (17.48 %) partly mediated the effect of WHtR on T2D in men. For women, this association was partly mediated by SBP (13.86 %), HDL (24.54 %), ALT (6.29 %), UA (22.58 %) and combined kidney indicators (39.51 %). CONCLUSIONS WHtR was an independent risk factor for the development of T2D and IFG. This association was partly mediated by HOMA-IR, SBP, lipids and other liver and kidney metabolism indicators.
Collapse
Affiliation(s)
- Ning Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Li-Kun Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yue Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Jing Dong
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xi Chu
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ya-Ke Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yu-Hong Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Lin-Lin Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| |
Collapse
|
22
|
McEvoy CM, Murphy JM, Zhang L, Clotet-Freixas S, Mathews JA, An J, Karimzadeh M, Pouyabahar D, Su S, Zaslaver O, Röst H, Arambewela R, Liu LY, Zhang S, Lawson KA, Finelli A, Wang B, MacParland SA, Bader GD, Konvalinka A, Crome SQ. Single-cell profiling of healthy human kidney reveals features of sex-based transcriptional programs and tissue-specific immunity. Nat Commun 2022; 13:7634. [PMID: 36496458 PMCID: PMC9741629 DOI: 10.1038/s41467-022-35297-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Knowledge of the transcriptional programs underpinning the functions of human kidney cell populations at homeostasis is limited. We present a single-cell perspective of healthy human kidney from 19 living donors, with equal contribution from males and females, profiling the transcriptome of 27677 cells to map human kidney at high resolution. Sex-based differences in gene expression within proximal tubular cells were observed, specifically, increased anti-oxidant metallothionein genes in females and aerobic metabolism-related genes in males. Functional differences in metabolism were confirmed in proximal tubular cells, with male cells exhibiting higher oxidative phosphorylation and higher levels of energy precursor metabolites. We identified kidney-specific lymphocyte populations with unique transcriptional profiles indicative of kidney-adapted functions. Significant heterogeneity in myeloid cells was observed, with a MRC1+LYVE1+FOLR2+C1QC+ population representing a predominant population in healthy kidney. This study provides a detailed cellular map of healthy human kidney, and explores the complexity of parenchymal and kidney-resident immune cells.
Collapse
Affiliation(s)
- Caitriona M McEvoy
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada
| | - Julia M Murphy
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lin Zhang
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Sergi Clotet-Freixas
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Jessica A Mathews
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - James An
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Mehran Karimzadeh
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
| | - Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Shenghui Su
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Olga Zaslaver
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Hannes Röst
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Rangi Arambewela
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Lewis Y Liu
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sally Zhang
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Keith A Lawson
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Antonio Finelli
- Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Bo Wang
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sonya A MacParland
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ana Konvalinka
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, ON, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Sarah Q Crome
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
23
|
Wang AC, Geng JH, Wang CW, Wu DW, Chen SC. Sex difference in the associations among risk factors with hepatitis B and C infections in a large Taiwanese population study. Front Public Health 2022; 10:1068078. [PMID: 36530675 PMCID: PMC9748294 DOI: 10.3389/fpubh.2022.1068078] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
Background The prevalence rates of hepatitis B and C virus (HBV/HCV) infection are high in Taiwan, and both are common causes of chronic liver disease and its related complications. Therefore, the early detection of factors associated with HBV/HCV infection is important. The aim of this study was to explore these factors in a large cohort of Taiwanese participants in the Taiwan Biobank, and also to identify sex differences in these risk factors. Methods It was an observational cohort study. The study enrolled 121,421 participants, and divided into four groups according to the presence or absence of HBV or HCV infection. Associations between risk factors with HBV or HCV infection were examined using multivariate logistic regression analysis. Results The mean age of the 121,421 enrolled participants (43,636 men and 77,785 women) was 49.9 ± 11.0 years. The participants were stratified into four groups according to those with (n = 13,804; 11.4%) and without HBV infection (n = 107,617; 88.6%), and those with (n = 2,750; 2.3%) and without HCV infection (n = 118,671; 97.7%). Multivariable analysis revealed that male sex [vs. female sex; odds ratio [OR] = 1.346; 95% confidence interval (CI) = 1.282-1.414; p < 0.001] was significantly associated with HBV infection, whereas female sex (vs. male sex; OR = 0.642; 95% CI = 0.575-0.716; p < 0.001) was significantly associated with HCV infection. Furthermore, there were significant interactions between sex and age (p < 0.001), body mass index (p < 0.001), total cholesterol (p = 0.002), aspartate aminotransferase (p = 0.024), and estimated glomerular filtration rate (p = 0.012) on HBV infection. There were also significant interactions between sex and age (p < 0.001), hypertension (p = 0.010), fasting glucose (p = 0.031), and uric acid (p = 0.001) on HCV infection. Conclusion In conclusion, sex differences were found among the risk factors for HBV and HCV infections in a large cohort of Taiwanese volunteers. When dealing with hepatitis B and hepatitis C, the physicians may need to pay attention to the differences between men and women to do different treatments.
Collapse
Affiliation(s)
- Angela Chiunhsien Wang
- Department of Post Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jiun-Hung Geng
- Department of Urology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Wen Wang
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Da-Wei Wu
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Szu-Chia Chen
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan,Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan,*Correspondence: Szu-Chia Chen
| |
Collapse
|
24
|
Park JW, Noh JH, Kim JM, Lee HY, Kim KA, Park JY. Gene Dose-Dependent and Additive Effects of ABCG2 rs2231142 and SLC2A9 rs3733591 Genetic Polymorphisms on Serum Uric Acid Levels. Metabolites 2022; 12:metabo12121192. [PMID: 36557230 PMCID: PMC9781553 DOI: 10.3390/metabo12121192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
This study aimed to evaluate whether the single nucleotide polymorphisms of ATP-binding cassette subfamily G member 2 (ABCG2) and solute carrier family 2 member 9 (SLC2A9) affect individual blood uric acid levels using pyrosequencing. ABCG2 (rs2231142, rs72552713, rs2231137), SLC2A9 (rs3734553, rs3733591, rs16890979), and individual uric acid levels were prospectively analyzed in 250 healthy young Korean male participants. Prominent differences in uric acid levels of the alleles were observed in the SLC2A9 rs3733591 polymorphism: wild-type (AA) vs. heterozygote (AG), 0.7 mg/dL (p < 0.0001); AA vs. mutant type (GG), 1.32 mg/dL (p < 0.0001); and AG vs. GG, 0.62 mg/dL (p < 0.01). In ABCG2 single nucleotide polymorphisms (SNPs), the statistically significant differences in uric acid levels were only found in rs2231142 between CC vs. AA (1.06 mg/dL; p < 0.001), and CC vs. CA (0.59 mg/dL; p < 0.01). Serum uric acid levels based on the ABCG2 and SLC2A9 diplotype groups were also compared. The uric acid levels were the lowest in the CC/AA diplotype and highest in the AA/AG diplotype. In addition, the SNP SLC2A9 rs3733591 tended to increase the uric acid levels when the ABCG2 rs2231142 haplotypes were fixed. In conclusion, both the ABCG2 rs2231142 and SLC2A9 rs3733591 polymorphisms may additively elevate blood uric acid levels.
Collapse
Affiliation(s)
- Jin-Woo Park
- Department of Clinical Pharmacology and Toxicology, Korea University Anam Hospital, Korea University Medicine, Seoul 02841, Republic of Korea
- Department of Neurology, Korea University Anam Hospital, Korea University Medicine, Seoul 02841, Republic of Korea
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Ji-Hyeon Noh
- Department of Clinical Pharmacology and Toxicology, Korea University Anam Hospital, Korea University Medicine, Seoul 02841, Republic of Korea
| | - Jong-Min Kim
- Department of Clinical Pharmacology and Toxicology, Korea University Anam Hospital, Korea University Medicine, Seoul 02841, Republic of Korea
| | - Hwa-Young Lee
- Department of Clinical Pharmacology and Toxicology, Korea University Anam Hospital, Korea University Medicine, Seoul 02841, Republic of Korea
| | - Kyoung-Ah Kim
- Department of Clinical Pharmacology and Toxicology, Korea University Anam Hospital, Korea University Medicine, Seoul 02841, Republic of Korea
| | - Ji-Young Park
- Department of Clinical Pharmacology and Toxicology, Korea University Anam Hospital, Korea University Medicine, Seoul 02841, Republic of Korea
- Correspondence: ; Tel.: +82-2-920-6288
| |
Collapse
|
25
|
Yang B, Xin M, Liang S, Xu X, Cai T, Dong L, Wang C, Wang M, Cui Y, Song X, Sun J, Sun W. New insight into the management of renal excretion and hyperuricemia: Potential therapeutic strategies with natural bioactive compounds. Front Pharmacol 2022; 13:1026246. [PMID: 36483739 PMCID: PMC9723165 DOI: 10.3389/fphar.2022.1026246] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/26/2022] [Indexed: 10/05/2023] Open
Abstract
Hyperuricemia is the result of increased production and/or underexcretion of uric acid. Hyperuricemia has been epidemiologically associated with multiple comorbidities, including metabolic syndrome, gout with long-term systemic inflammation, chronic kidney disease, urolithiasis, cardiovascular disease, hypertension, rheumatoid arthritis, dyslipidemia, diabetes/insulin resistance and increased oxidative stress. Dysregulation of xanthine oxidoreductase (XOD), the enzyme that catalyzes uric acid biosynthesis primarily in the liver, and urate transporters that reabsorb urate in the renal proximal tubules (URAT1, GLUT9, OAT4 and OAT10) and secrete urate (ABCG2, OAT1, OAT3, NPT1, and NPT4) in the renal tubules and intestine, is a major cause of hyperuricemia, along with variations in the genes encoding these proteins. The first-line therapeutic drugs used to lower serum uric acid levels include XOD inhibitors that limit uric acid biosynthesis and uricosurics that decrease urate reabsorption in the renal proximal tubules and increase urate excretion into the urine and intestine via urate transporters. However, long-term use of high doses of these drugs induces acute kidney disease, chronic kidney disease and liver toxicity. Therefore, there is an urgent need for new nephroprotective drugs with improved safety profiles and tolerance. The current systematic review summarizes the characteristics of major urate transporters, the mechanisms underlying the pathogenesis of hyperuricemia, and the regulation of uric acid biosynthesis and transport. Most importantly, this review highlights the potential mechanisms of action of some naturally occurring bioactive compounds with antihyperuricemic and nephroprotective potential isolated from various medicinal plants.
Collapse
Affiliation(s)
- Bendong Yang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Meiling Xin
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Shufei Liang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Xiaoxue Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Tianqi Cai
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Ling Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Chao Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Meng Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Xinhua Song
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
- Shandong Qingyujiangxing Biotechnology Co., Ltd., Zibo, China
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Wenlong Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
- Shandong Qingyujiangxing Biotechnology Co., Ltd., Zibo, China
| |
Collapse
|
26
|
Chang YS, Lin CY, Liu TY, Huang CM, Chung CC, Chen YC, Tsai FJ, Chang JG, Chang SJ. Polygenic risk score trend and new variants on chromosome 1 are associated with male gout in genome-wide association study. Arthritis Res Ther 2022; 24:229. [PMID: 36221101 PMCID: PMC9552457 DOI: 10.1186/s13075-022-02917-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 09/24/2022] [Indexed: 11/30/2022] Open
Abstract
Background Gout is a highly hereditary disease, but not all those carrying well-known risk variants have developing gout attack even in hyperuricemia status. We performed a genome-wide association study (GWAS) and polygenic risk score (PRS) analysis to illustrate the new genetic architectures of gout and asymptomatic hyperuricemia (AH). Methods GWAS was performed to identify variants associated with gout/AH compared with normouricemia. The participants were males, enrolled from the Taiwan Biobank and China Medical University, and divided into discovery (n=39,594) and replication (n=891) cohorts for GWAS. For PRS analysis, the discovery cohort was grouped as base (n=21,814) and target (n=17,780) cohorts, and the score was estimated by grouping the polymorphisms into protective or not for the phenotypes in the base cohort. Results The genes ABCG2 and SLC2A9 were found as the major genetic factors governing gouty and AH, and even in those carrying the rs2231142 (ABCG2) wild-genotype. Surprisingly, variants on chromosome 1, such as rs7546668 (DNAJC16), rs10927807 (AGMAT), rs9286836 (NUDT17), rs4971100 (TRIM46), rs4072037 (MUC1), and rs2974935 (MTX1), showed significant associations with gout in both discovery and replication cohorts (all p-values < 1e−8). Concerning the PRS, the rates of gout and AH increased with increased quartile PRS in those SNPs having risk effects on the phenotypes; on the contrary, gout/AH rates decreased with increased quartile PRS in those protective SNPs. Conclusions We found new variants on chromosome 1 significantly relating to gout, and PRS predicts the risk of developing gout/AH more robustly based on the SNPs’ effect types on the trait. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02917-4.
Collapse
Affiliation(s)
- Ya-Sian Chang
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chien-Yu Lin
- Graduate Institute of Clinical Medical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Division of Laboratory Medicine, China Medical University Hsinchu Hospital, Zhubei City, Taiwan
| | - Ting-Yuan Liu
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chung-Ming Huang
- Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chin-Chun Chung
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Chia Chen
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| | - Jan-Gowth Chang
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Graduate Institute of Integrated Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| | - Shun-Jen Chang
- Center for Precision Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, No. 700, Kaohsiung University Road, Nanzih District, 81148, Kaohsiung, Taiwan.
| |
Collapse
|
27
|
Yong T, Liang D, Chen S, Xiao C, Gao X, Wu Q, Xie Y, Huang L, Hu H, Li X, Liu Y, Cai M. Caffeic acid phenethyl ester alleviated hypouricemia in hyperuricemic mice through inhibiting XOD and up-regulating OAT3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154256. [PMID: 35714456 DOI: 10.1016/j.phymed.2022.154256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Hyperuricemia is characterized with high serum uric acids (SUAs) and directly causes suffering gout. Caffeic acid phenethyl ester (CAPE) is widely included in dietary plants and especially propolis of honey hives. HYPOTHESIS/PURPOSE Since CAPE exerts a property resembling a redox shuttle, the hypothesis is that it may suppress xanthine oxidase (XOD) and alleviate hyperuricemia. The aim is to unveil the hypouricemic effect of CAPE and the underlying mechanisms. METHODS By establishing a hyperuricemic model with potassium oxonate (PO) and hypoxanthine (HX) together, we investigated the hypouricecmic effect of CAPE. On this model, the expressions of key mRNAs and proteins, including glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1), and the activity of XOD were assayed in vivo. Also, the inhibitory effect of CAPE against XOD was assayed in vitro through enzymatic activity tests and by molecular docking. RESULTS CAPE demonstrated a remarkable hypouricemic effect, which reduced the SUAs of hyperuricemic mice (401 ± 111 µmol/l) to 209 ± 56, 204 ± 65 and 154 ± 40 µmol/l (p < 0.01) at the doses of 15, 30 and 60 mg/kg respectively, depicting efficacies between 48 and 62% and approaching allopurinol's efficacy (52%). Serum parameters, body weights, inner organ coefficients, and H&E staining suggested that CAPE displayed no general toxicity and it alleviated the liver and kidney injuries caused by hyperuricemia. Mechanistically, CAPE decreased XOD activities significantly in vivo, presented an IC50 at 214.57 µM in vitro and depicted a favorable binding to XOD in molecular simulation, indicating that inhibiting XOD may be an underlying mechanism of CAPE against hyperuricemia. CAPE did decreased GLUT9 protein and down-regulated URAT1 mRNA and protein. In addition, CAPE up-regulated ATP binding cassette subfamily G member 2 (ABCG2) and organic anion transporter 3 (OAT3) mRNA and proteins in comparison with that of the hyperuricemic control. All above, CAPE may alleviate hyperuricmia through inhibiting XOD, decreasing GLUT9 and URAT1 and increasing ABCG2 and OAT3. CONCLUSION CAPE presented potent hypouricemic effect in hyperuricemic mice through inhibiting XOD activity and up-regulating OAT3. CAPE may be a promising treatment against hyperuricemia.
Collapse
Affiliation(s)
- Tianqiao Yong
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Danling Liang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Shaodan Chen
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Chun Xiao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Xiong Gao
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China.
| | - Yizhen Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Longhua Huang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Huiping Hu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Xiangmin Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yuancao Liu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Manjun Cai
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, Key Laboratory of Agricultural Microbiomics and Precision Application of the Ministry of Agriculture and Rural Affairs and State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| |
Collapse
|
28
|
Zhao H, Lu Z, Lu Y. The potential of probiotics in the amelioration of hyperuricemia. Food Funct 2022; 13:2394-2414. [PMID: 35156670 DOI: 10.1039/d1fo03206b] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hyperuricemia is a common disease caused by metabolic disorders or the excessive intake of high-purine foods. Persistent hyperuricemia in extreme cases induces gout, and asymptomatic hyperuricemia is probably linked to other metabolic diseases, such as hypertension. The typical damage caused by asymptomatic hyperuricemia includes inflammation, oxidative stress and gut dysbiosis. Probiotics have broad potential applications as food additives, not as drug therapies, in the amelioration of hyperuricemia. In this review, we describe novel methods for potential hyperuricemia amelioration with probiotics. The pathways through which probiotics may ameliorate hyperuricemia are discussed, including the decrease in uric acid production through purine assimilation and XOD (xanthine oxidase) inhibition as well as enhanced excretion of uric acid production by promoting ABCG2 (ATP binding cassette subfamily G member 2) activity, respectively. Three possible probiotic-related therapeutic pathways for alleviating the syndrome of hyperuricemia are also summarized. The first mechanism is to alleviate the oxidation and inflammation induced by hyperuricemia through the inhibition of NLRP3 inflammasome, the second is to restore damaged intestinal epithelium barriers and prevent gut microbiota dysbiosis, and the third is to enhance the innate immune system by increasing the secretion of immunoglobulin A (sIgA) to resist the stimulus by hyperuricemia. We propose that future research should focus on superior strain resource isolation and insight into the cause-effect mechanisms of probiotics for hyperuricemia amelioration. The safety and effects of the application of probiotics in clinical use also need verification.
Collapse
Affiliation(s)
- Hongyuan Zhao
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Zhaoxin Lu
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yingjian Lu
- College of Food Science & Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China.
| |
Collapse
|
29
|
Serum uric acid level is associated with an increase in systolic blood pressure over time in female subjects: Linear mixed-effects model analyses. Hypertens Res 2022; 45:344-353. [PMID: 34848887 DOI: 10.1038/s41440-021-00792-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Whether hyperuricemia is a true risk factor for elevated blood pressure (BP) is controversial, and the sex-specific effects of serum uric acid (SUA) on BP during a follow-up period remain unclear. We investigated whether the association of SUA level with systolic or diastolic BP during a 10-year period differs by sex in a Japanese general population of individuals who received annual health examinations (n = 28,990). After exclusion of subjects who had no BP or SUA data at baseline, a total of 22,994 subjects (male/female: 14,603/8391, age: 47 ± 11 years) were recruited. After adjustment for age; body mass index; BP; SUA level; use of drugs for hyperuricemia and hypertension; diagnosis of diabetes mellitus, dyslipidemia, and chronic kidney disease; family history of hypertension; habits of current smoking and alcohol consumption at baseline; the duration of the observation period; and the interaction between each covariate and the duration of the observation period indicated a significant association of SUA level with change in systolic or diastolic BP over time. There was a significant interaction between sex and SUA level for the change in systolic BP (P = 0.003) but not the change in diastolic BP (P = 0.081). The SUA level at baseline (per 1 mg/dL) was significantly associated with a change in systolic BP over time in females (estimate: 0.073 mmHg/year, P = 0.003) but not in males (estimate: 0.020 mmHg/year, P = 0.160). In conclusion, a high SUA level at baseline is significantly associated with an increase in systolic BP over time in female individuals but not in male individuals.
Collapse
|
30
|
Cheng F, Li Y, Zheng H, Tian L, Jia H. Mediating Effect of Body Mass Index and Dyslipidemia on the Relation of Uric Acid and Type 2 Diabetes: Results From China Health and Retirement Longitudinal Study. Front Public Health 2022; 9:823739. [PMID: 35155363 PMCID: PMC8831836 DOI: 10.3389/fpubh.2021.823739] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 12/28/2021] [Indexed: 12/15/2022] Open
Abstract
ObjectiveThis study assessed temporal relationships of serum uric acid (SUA) with blood glucose and determine the mediating effects of body mass index (BMI) and dyslipidemia on the relation of SUA and risk of type 2 diabetes.MethodsParticipants aged ≥ 45 years were participated in 2011 and followed up until 2015. Cox proportional hazards regression with a robust variance estimator was performed to explore the association of SUA with the risk of diabetes, and crosslagged path analysis was introduced to examine the temporal relationships between SUA and blood glucose. A mediation analysis was finally used to identify the mediating effect of BMI and dyslipidemia on the relation of SUA and the future risk of diabetes.ResultsA total of 9,020 participants were included with an average age of 58.59 years at baseline in 2011, and 53.6% of them were women. Linear dose–response relationship was identified by restricted spline cubic analysis between baseline SUA and follow-up blood glucose (the non-linear trend for fasting plasma glucose (FPG): β2 = −0.71, p = 0.52; for HbA1c: β2 = 0.05, p = 0.07; for risk of diabetes: β2 = 0.12, p = 0.39). Additionally, compared with the lowest quartiles of SUA, the adjusted risk ratios of diabetes were 1.00 (95% CI: 0.82–1.23), 1.08 (95% CI: 0.89–1.31), and 1.37 (95% CI: 1.11–1.96) for quartile 2–4 (p-trend < 0.01), respectively. Further additional adjustments for BMI or dyslipidemia, these ratios were not statistically significant. In addition, a unidirectional relationship from baseline SUA to follow-up FPG (ρ1 = 0.24, p = 0.03) was further confirmed using crosslagged path analysis. After stratifying by genders, the above results were only significant in the women subgroup, and we thus conducted a mediation analysis in women and found that the BMI and dyslipidemia partially mediated the effect of SUA on diabetes with a 23.05 and 18.82% mediating effect, respectively.ConclusionsThese findings provide strong evidence that hyperuricemia preceded diabetes, and the effect of baseline SUA on follow-up type 2 diabetes was more pronounced among middle-aged and elderly Chinese women, especially in postmenopausal women, and this effect is partly mediated by BMI and dyslipidemia at baseline.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Center of Evidence-Based Medicine, Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanzhi Li
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Han Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lu Tian
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongying Jia
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Center of Evidence-Based Medicine, Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Hongying Jia
| |
Collapse
|
31
|
Association and interaction between dietary patterns and gene polymorphisms in Liangshan residents with hyperuricemia. Sci Rep 2022; 12:1356. [PMID: 35079028 PMCID: PMC8789849 DOI: 10.1038/s41598-021-04568-y] [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: 06/14/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Hyperuricemia (HUA) is associated with dietary and genetic factors. However, studies on dietary patterns and their interaction effect with genes on the risk of HUA are limited. We aimed to explore the association between dietary patterns and HUA, and dietary patterns—gene interactions on the risk of HUA. A population-based cross-sectional study was conducted in adults aged 18 and older in Liangshan Yi Autonomous Prefecture of China. Dietary consumption was collected using a standard Food Frequency Questionnaire. Vein blood samples were collected after overnight fasting, and DNA was extracted from peripheral blood leukocytes. Dietary patterns were derived using principal component and factor analysis. Of the 2646 participants, the prevalence of HUA was 26.8%. Three dietary patterns were classified. Of them, a dietary pattern with higher meat consumption (defined as meat-based) had the strongest association with HUA than a dietary pattern with plant-based or local special diet-based. A higher frequency of T allele at ABCG2 rs2231142 and SLC2A9 rs11722228 loci was observed in participants with HUA than those without HUA. An additive interaction of meat-based dietary pattern with rs2231142 locus was significantly associated with an increased risk of HUA. The relative excess risks of interaction, attributable proportion of interaction, and synergy index (S) were 0.482 (95% CI: 0.012–0.976), 0.203 (95% CI: 0.033–0.374), and 1.544 (95% CI: 1.012–2.355), respectively. In conclusion, a dietary pattern with meat-based was significantly associated with an increased risk of HUA. There was an additive interaction between a meat-based dietary pattern and the ABCG2 rs2231142 locus. Individuals with rs2231142 T allele were at higher risk of HUA than those with rs2231142 GG allele.
Collapse
|
32
|
Chen Y, Yang Y, Zhong Y, Li J, Kong T, Zhang S, Yang S, Wu C, Cui B, Fu L, Hui R, Zhang W. Genetic risk of hyperuricemia in hypertensive patients associated with antihypertensive drug therapy: a longitudinal study. Clin Genet 2022; 101:411-420. [PMID: 35023146 PMCID: PMC9306909 DOI: 10.1111/cge.14110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/25/2021] [Accepted: 01/10/2022] [Indexed: 11/27/2022]
Abstract
Elevated serum uric acid (UA) level has been shown to be influenced by multiple genetic variants, but it remains uncertain how UA‐associated variants differ in their influence on hyperuricemia risk in people taking antihypertensive drugs. We examined a total of 43 UA‐related variants at 29 genes in 1840 patients with hypertension from a community‐based longitudinal cohort during a median 2.25‐year follow‐up (including 1031 participants with normal UA, 440 prevalent hyperuricemia at baseline, and 369 new‐onset hyperuricemia). Compared with the wild‐type genotypes, patients carrying the SLC2A9 rs3775948G allele or the rs13129697G allele had decreased risk of hyperuricemia, while patients carrying the SLC2A9 rs11722228T allele had increased risk of hyperuricemia, after adjustment for cardiovascular risk factors and correction for multiple comparisons; moreover, these associations were modified by the use of diuretics, β‐blockers, or angiotensin converting enzyme inhibitors. The rs10821905A allele of A1CF gene was associated with increased risk of hyperuricemia, and this risk was enhanced by diuretics use. The studied variants were not observed to confer risk for incident cardiovascular events during the follow‐up. In conclusion, the genes SLC2A9 and A1CF may serve as potential genetic markers for hyperuricemia risk in relation to antihypertensive drugs therapy in Chinese hypertensive patients.
Collapse
Affiliation(s)
- Yu Chen
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yunyun Yang
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yixuan Zhong
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Li
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Kong
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Shuyuan Zhang
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Shujun Yang
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Cunjin Wu
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bing Cui
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Li Fu
- Benxi Railway Hospital, Benxi, China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Weili Zhang
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Zhang X, Hou G, Li F, Zheng X, Nie Q, Song G. SLC2A9 rs1014290 Polymorphism is Associated with Prediabetes and Type 2 Diabetes. Int J Endocrinol 2022; 2022:4947684. [PMID: 36545489 PMCID: PMC9763018 DOI: 10.1155/2022/4947684] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/17/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE To investigate the association of the A/G rs1014290 polymorphism in SLC2A9 with type 2 diabetes (T2DM) and prediabetes mellitus (pre-DM). Patients and Methods. We enrolled 1058 patients who attended the Hebei General Hospital, Shijiazhuang, Hebei Province, China. The patients underwent general testing and oral glucose tolerance tests and were divided into three groups: 352 patients newly diagnosed with T2DM, 358 patients with pre-DM, and 348 healthy controls. The single nucleotide polymorphism (SNP) was detected by ligase detection reactions. The χ 2 test, one-way ANOVA, and binary logistic regression analysis were used to analyze the results. RESULTS In the T2DM group, the GG genotype frequency at the rs1014290 locus was significantly lower (14.8%) than it was in the healthy controls. Furthermore, the GG genotype group was associated with a reduced risk of T2DM in unadjusted and confounder-adjusted models compared with the risk in the AA genotype group. The G allele in the SLC2A9 rs1014290 locus decreased susceptibility to T2DM. In the pre-DM group, the GG and AG genotype groups had no significant correlation with the risk of pre-DM in any of the models. In the T2DM group, the uric acid level was significantly lower in the GG genotype group. In the T2DM and pre-DM groups, the HOMA-β levels were significantly higher in the GA (P < 0.001) and GG (P < 0.001) genotype groups than it was in the AA genotype group, and HOMA-IR was significantly lower in the GA (P < 0.001) and GG (P < 0.001) genotype groups than it was in the AA genotype group. CONCLUSION The A/G (rs1014290) SNP in SLC2A9 is closely related to the occurrence and development of diabetes.
Collapse
Affiliation(s)
- Xuemei Zhang
- Department of Rheumatism and Immunology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Guangsen Hou
- Department of Geriatric, Affiliated Hospital of Hebei Engineering University, 81 Congtai Road, Handan, Hebei 056000, China
| | - Fang Li
- Department of Rheumatism and Immunology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Xiao Zheng
- Department of Rheumatism and Immunology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Qian Nie
- Physical Examination Center, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, Hebei 050000, China
| | - Guangyao Song
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
34
|
Yang Y, Xian W, Wu D, Huo Z, Hong S, Li Y, Xiao H. The role of obesity, type 2 diabetes, and metabolic factors in gout: A Mendelian randomization study. Front Endocrinol (Lausanne) 2022; 13:917056. [PMID: 35992130 PMCID: PMC9388832 DOI: 10.3389/fendo.2022.917056] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Several epidemiological studies have reported a possible correlation between risk of gout and metabolic disorders including type 2 diabetes, insulin resistance, obesity, dyslipidemia, and hypertension. However, it is unclear if this association is causal. METHODS We used Mendelian randomization (MR) to evaluate the causal relation between metabolic conditions and gout or serum urate concentration by inverse-variance-weighted (conventional) and weighted median methods. Furthermore, MR-Egger regression and MR-pleiotropy residual sum and outlier (PRESSO) method were used to explore pleiotropy. Genetic instruments for metabolic disorders and outcome (gout and serum urate) were obtained from several genome-wide association studies on individuals of mainly European ancestry. RESULTS Conventional MR analysis showed a robust causal association of increasing obesity measured by body mass index (BMI), high-density lipoprotein cholesterol (HDL), and systolic blood pressure (SBP) with risk of gout. A causal relationship between fasting insulin, BMI, HDL, triglycerides (TG), SBP, alanine aminotransferase (ALT), and serum urate was also observed. These results were consistent in weighted median method and MR-PRESSO after removing outliers identified. Our analysis also indicated that HDL and serum urate as well as gout have a bidirectional causal effect on each other. CONCLUSIONS Our study suggested causal effects between glycemic traits, obesity, dyslipidemia, blood pressure, liver function, and serum urate as well as gout, which implies that metabolic factors contribute to the development of gout via serum urate, as well as potential benefit of sound management of increased serum urate in patients with obesity, dyslipidemia, hypertension, and liver dysfunction.
Collapse
|
35
|
Sędzikowska A, Szablewski L. Human Glucose Transporters in Renal Glucose Homeostasis. Int J Mol Sci 2021; 22:13522. [PMID: 34948317 PMCID: PMC8708129 DOI: 10.3390/ijms222413522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022] Open
Abstract
The kidney plays an important role in glucose homeostasis by releasing glucose into the blood stream to prevent hypoglycemia. It is also responsible for the filtration and subsequent reabsorption or excretion of glucose. As glucose is hydrophilic and soluble in water, it is unable to pass through the lipid bilayer on its own; therefore, transport takes place using carrier proteins localized to the plasma membrane. Both sodium-independent glucose transporters (GLUT proteins) and sodium-dependent glucose transporters (SGLT proteins) are expressed in kidney tissue, and mutations of the genes coding for these glucose transporters lead to renal disorders and diseases, including renal cancers. In addition, several diseases may disturb the expression and/or function of renal glucose transporters. The aim of this review is to describe the role of the kidney in glucose homeostasis and the contribution of glucose transporters in renal physiology and renal diseases.
Collapse
Affiliation(s)
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland;
| |
Collapse
|
36
|
Tin A, Schlosser P, Matias-Garcia PR, Thio CHL, Joehanes R, Liu H, Yu Z, Weihs A, Hoppmann A, Grundner-Culemann F, Min JL, Kuhns VLH, Adeyemo AA, Agyemang C, Ärnlöv J, Aziz NA, Baccarelli A, Bochud M, Brenner H, Bressler J, Breteler MMB, Carmeli C, Chaker L, Coresh J, Corre T, Correa A, Cox SR, Delgado GE, Eckardt KU, Ekici AB, Endlich K, Floyd JS, Fraszczyk E, Gao X, Gào X, Gelber AC, Ghanbari M, Ghasemi S, Gieger C, Greenland P, Grove ML, Harris SE, Hemani G, Henneman P, Herder C, Horvath S, Hou L, Hurme MA, Hwang SJ, Kardia SLR, Kasela S, Kleber ME, Koenig W, Kooner JS, Kronenberg F, Kühnel B, Ladd-Acosta C, Lehtimäki T, Lind L, Liu D, Lloyd-Jones DM, Lorkowski S, Lu AT, Marioni RE, März W, McCartney DL, Meeks KAC, Milani L, Mishra PP, Nauck M, Nowak C, Peters A, Prokisch H, Psaty BM, Raitakari OT, Ratliff SM, Reiner AP, Schöttker B, Schwartz J, Sedaghat S, Smith JA, Sotoodehnia N, Stocker HR, Stringhini S, Sundström J, Swenson BR, van Meurs JBJ, van Vliet-Ostaptchouk JV, Venema A, Völker U, Winkelmann J, Wolffenbuttel BHR, Zhao W, Zheng Y, Loh M, Snieder H, Waldenberger M, Levy D, Akilesh S, Woodward OM, Susztak K, Teumer A, Köttgen A. Epigenome-wide association study of serum urate reveals insights into urate co-regulation and the SLC2A9 locus. Nat Commun 2021; 12:7173. [PMID: 34887389 PMCID: PMC8660809 DOI: 10.1038/s41467-021-27198-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022] Open
Abstract
Elevated serum urate levels, a complex trait and major risk factor for incident gout, are correlated with cardiometabolic traits via incompletely understood mechanisms. DNA methylation in whole blood captures genetic and environmental influences and is assessed in transethnic meta-analysis of epigenome-wide association studies (EWAS) of serum urate (discovery, n = 12,474, replication, n = 5522). The 100 replicated, epigenome-wide significant (p < 1.1E-7) CpGs explain 11.6% of the serum urate variance. At SLC2A9, the serum urate locus with the largest effect in genome-wide association studies (GWAS), five CpGs are associated with SLC2A9 gene expression. Four CpGs at SLC2A9 have significant causal effects on serum urate levels and/or gout, and two of these partly mediate the effects of urate-associated GWAS variants. In other genes, including SLC7A11 and PHGDH, 17 urate-associated CpGs are associated with conditions defining metabolic syndrome, suggesting that these CpGs may represent a blood DNA methylation signature of cardiometabolic risk factors. This study demonstrates that EWAS can provide new insights into GWAS loci and the correlation of serum urate with other complex traits.
Collapse
Affiliation(s)
- Adrienne Tin
- Department of Medicine, University of Mississippi Medical Center, Jackson, 39216, MS, USA.
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Pascal Schlosser
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Pamela R Matias-Garcia
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
- TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Chris H L Thio
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Roby Joehanes
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hongbo Liu
- Department of Medicine and Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Zhi Yu
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Antoine Weihs
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Anselm Hoppmann
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Franziska Grundner-Culemann
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Josine L Min
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | | | - Adebowale A Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles Agyemang
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society (NVS), Family Medicine and Primary Care Unit, Karolinska Institutet, Huddinge, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| | - Nasir A Aziz
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Andrea Baccarelli
- Laboratory of Environmental Precision Health, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Murielle Bochud
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Hermann Brenner
- German Cancer Research Center (DKFZ), Division of Clinical Epidemiology and Aging Research, Heidelberg, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Bressler
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, 77030, TX, USA
| | - Monique M B Breteler
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Cristian Carmeli
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
- Population Health Laboratory, University of Fribourg, Fribourg, Switzerland
| | - Layal Chaker
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Tanguy Corre
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, 39216, MS, USA
| | - Simon R Cox
- Lothian Birth Cohorts Group, Department of Psychology, The University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Graciela E Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander-UniversitätErlangen-Nürnberg, 91054, Erlangen, Germany
| | - Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - James S Floyd
- Department of Medicine, University of Washington, Seattle, 98101, WA, USA
- Department of Epidemiology, University of Washington, Seattle, 98101, WA, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, 98101, WA, USA
| | - Eliza Fraszczyk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Xu Gao
- Laboratory of Environmental Precision Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Xīn Gào
- German Cancer Research Center (DKFZ), Division of Clinical Epidemiology and Aging Research, Heidelberg, Germany
| | - Allan C Gelber
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sahar Ghasemi
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
| | - Philip Greenland
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, 77030, TX, USA
| | - Sarah E Harris
- Lothian Birth Cohorts Group, Department of Psychology, The University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Peter Henneman
- Department of Clinical Genetics, Amsterdam Reproduction & Development Research Institute, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Munich-Neuherberg, Germany
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, 90095, CA, USA
- Biostatistics, Fielding School of Public Health, UCLA, Los Angeles, CA, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mikko A Hurme
- Department of Microbiology and Immunology, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Shih-Jen Hwang
- Framingham Heart Study, Framingham, MA, USA
- Division of Intramural Research, Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Silva Kasela
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Marcus E Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm, Germany
| | - Jaspal S Kooner
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, Southall, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Brigitte Kühnel
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
| | - Christine Ladd-Acosta
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Centre, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Dan Liu
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Ake T Lu
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, 90095, CA, USA
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Karlijn A C Meeks
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Centre, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Matthias Nauck
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society (NVS), Family Medicine and Primary Care Unit, Karolinska Institutet, Huddinge, Sweden
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
- Ludwig-Maximilians Universität München, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Department of Computational Health, Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
| | - Bruce M Psaty
- Department of Medicine, University of Washington, Seattle, 98101, WA, USA
- Department of Epidemiology, University of Washington, Seattle, 98101, WA, USA
- Cardiovascular Health Research Unit, University of Washington, Seattle, 98101, WA, USA
- Department of Health Services, University of Washington, Seattle, 98101, WA, USA
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Scott M Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Alex P Reiner
- Department of Epidemiology, University of Washington, Seattle, 98101, WA, USA
| | - Ben Schöttker
- German Cancer Research Center (DKFZ), Division of Clinical Epidemiology and Aging Research, Heidelberg, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, 48109, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, University of Washington, Seattle, 98101, WA, USA
| | - Hannah R Stocker
- German Cancer Research Center (DKFZ), Division of Clinical Epidemiology and Aging Research, Heidelberg, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Silvia Stringhini
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Brenton R Swenson
- Cardiovascular Health Research Unit, University of Washington, Seattle, 98101, WA, USA
- Institute for Public Health Genetics, University of Washington, Seattle, WA, USA
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jana V van Vliet-Ostaptchouk
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Andrea Venema
- Department of Clinical Genetics, Amsterdam Reproduction & Development Research Institute, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, the Netherlands
| | - Uwe Völker
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Juliane Winkelmann
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Chair Neurogenetics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, D-85764, Bavaria, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Katalin Susztak
- Department of Medicine and Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Alexander Teumer
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
37
|
Liu X, Tong X, Zhu J, Tian L, Jie Z, Zou Y, Lin X, Liang H, Li W, Ju Y, Qin Y, Zou L, Lu H, Zhu S, Jin X, Xu X, Yang H, Wang J, Zong Y, Liu W, Hou Y, Jia H, Zhang T. Metagenome-genome-wide association studies reveal human genetic impact on the oral microbiome. Cell Discov 2021; 7:117. [PMID: 34873157 PMCID: PMC8648780 DOI: 10.1038/s41421-021-00356-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/15/2021] [Indexed: 11/09/2022] Open
Abstract
The oral microbiota contains billions of microbial cells, which could contribute to diseases in many body sites. Challenged by eating, drinking, and dental hygiene on a daily basis, the oral microbiota is regarded as highly dynamic. Here, we report significant human genomic associations with the oral metagenome from more than 1915 individuals, for both the tongue dorsum (n = 2017) and saliva (n = 1915). We identified five genetic loci associated with oral microbiota at study-wide significance (p < 3.16 × 10-11). Four of the five associations were well replicated in an independent cohort of 1439 individuals: rs1196764 at APPL2 with Prevotella jejuni, Oribacterium uSGB 3339 and Solobacterium uSGB 315; rs3775944 at the serum uric acid transporter SLC2A9 with Oribacterium uSGB 1215, Oribacterium uSGB 489 and Lachnoanaerobaculum umeaense; rs4911713 near OR11H1 with species F0422 uSGB 392; and rs36186689 at LOC105371703 with Eggerthia. Further analyses confirmed 84% (386/455 for tongue dorsum) and 85% (391/466 for saliva) of host genome-microbiome associations including six genome-wide significant associations mutually validated between the two niches. As many of the oral microbiome-associated genetic variants lie near miRNA genes, we tentatively validated the potential of host miRNAs to modulate the growth of specific oral bacteria. Human genetics accounted for at least 10% of oral microbiome compositions between individuals. Machine learning models showed that polygenetic risk scores dominated over oral microbiome in predicting risk of dental diseases such as dental calculus and gingival bleeding. These findings indicate that human genetic differences are one explanation for a stable or recurrent oral microbiome in each individual.
Collapse
Affiliation(s)
- Xiaomin Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Xin Tong
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Jie Zhu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Liu Tian
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Zhuye Jie
- BGI-Shenzhen, Shenzhen, Guangdong, China
- Department of Biology, University of Copenhagen, Universitetsparken 13, Copenhagen, Denmark
| | - Yuanqiang Zou
- BGI-Shenzhen, Shenzhen, Guangdong, China
- Department of Biology, University of Copenhagen, Universitetsparken 13, Copenhagen, Denmark
- Qingdao-Europe Advanced Institute for Life Sciences, BGI-Shenzhen, Qingdao, Shandong, China
| | - Xiaoqian Lin
- BGI-Shenzhen, Shenzhen, Guangdong, China
- School of Bioscience and Biotechnology, South China University of Technology, Guangzhou, Guangdong, China
| | | | - Wenxi Li
- BGI-Shenzhen, Shenzhen, Guangdong, China
- School of Bioscience and Biotechnology, South China University of Technology, Guangzhou, Guangdong, China
| | - Yanmei Ju
- BGI-Shenzhen, Shenzhen, Guangdong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Youwen Qin
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Leying Zou
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Haorong Lu
- China National Genebank, BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Shida Zhu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, Guangdong, China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, Guangdong, China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, China
| | - Yang Zong
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Weibin Liu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yong Hou
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen, Guangdong, China.
| | - Tao Zhang
- BGI-Shenzhen, Shenzhen, Guangdong, China.
- Department of Biology, University of Copenhagen, Universitetsparken 13, Copenhagen, Denmark.
| |
Collapse
|
38
|
Zhang CL, Zhang JJ, Zhu QF, Guan HY, Yang YX, He X, Fu Y, Chen TX, Dong L, Yang XS, Tang KF, Xu GB, Liao SG. Antihyperuricemia and antigouty arthritis effects of Persicaria capitata herba in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153765. [PMID: 34610527 DOI: 10.1016/j.phymed.2021.153765] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Hyperuricemia (HUA) is an important risk factor for gout, renal dysfunction and cardiovascular diseases. The whole plant of Persicaria capitata (Buch.-Ham. ex D. Don) H. Gross, namely Persicaria capitata herba, is a well-known ethnic herb with potent therapeutic effects on urinary tract infections and urinary calculus, yet previous reports have only focused on its effect on urinary tract infections. PURPOSE To evaluate the therapeutic potential of P. capitata herba against gout by investigating its antihyperuricemia and antigouty arthritis effects and possible mechanisms. METHODS The ethanol extract (EP) and water extract (WP) of P. capitata herba were prepared by extracting dried and ground whole plants of P. capitata with 75% ethanol and water, respectively, followed by removal of solvents and characterization by UHPLC-Q-TOF/MS. The antihyperuricemia and antigouty arthritis effects of the two extracts were evaluated in a potassium oxonate- and hypoxanthine-induced hyperuricemia mouse model and a monosodium urate crystal (MSUC)-induced acute gouty arthritis mouse model, respectively. The mechanisms were investigated by testing their effects on the expression of correlated proteins (by Western blot) and mRNAs (by RT-PCR). RESULTS UHPLC-HRMS fingerprinting and two chemical markers (i.e., quercetin and quercitrin) determination were used for the characterization of the WP and EP extracts. Both WP and EP extracts showed pronounced antihyperuricemia activities, with a remarkable decline in serum uric acid and a marked increase in urine uric acid in hyperuricemic mice. Unlike the clinical xanthine oxidase (XOD) inhibitor allopurinol, WP and EP did not show any distinct renal toxicities. The underlying antihyperuricemia mechanism involves the inhibition of the activity and expression of XOD and the downregulation of the mRNA and protein expression of glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1). The extracts of P. capitata herba also demonstrated remarkable anti-inflammatory activity in MSUC-induced acute gouty arthritis mice. The mechanism might involve inhibitory effects on the expression of proinflammatory factors. CONCLUSIONS The extracts of P. capitata herba possessed pronounced antihyperuricemia and antigouty arthritis effects and were, therefore, promising natural medicines for hyperuricemia-related disorders and gouty arthritis. The use of P. capitata herba for the treatment of urinary calculus may be, at least to some degree, related to its potential as an antihyperuricemia and antigouty arthritis drug.
Collapse
Affiliation(s)
- Chun-Lei Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China.
| | - Jin-Juan Zhang
- School of Basic Medical Sciences, Guizhou Medical University, Guizhou 550025, China.
| | - Qin-Feng Zhu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China.
| | - Huan-Yu Guan
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China.
| | - Ya-Xin Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China.
| | - Xun He
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China.
| | - Yao Fu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China.
| | - Teng-Xiang Chen
- Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.
| | - Li Dong
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China.
| | - Xiao-Sheng Yang
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, Guizhou, China.
| | - Kai-Fa Tang
- Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.
| | - Guo-Bo Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China; National Engineering Research Center of Miao's Medicines & Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education & Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, 550004, Guizhou, China.
| | - Shang-Gao Liao
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 550025, Guizhou, China; National Engineering Research Center of Miao's Medicines & Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, Ministry of Education & Guizhou Provincial Key Laboratory of Pharmaceutics, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
39
|
Galvão AIR, Beleigoli AMR, Vidigal PG, Duncan BB, Schmidt MI, Appleton SL, Barreto SM, Diniz MDFHS. The positive association between serum uric acid, impaired fasting glucose, impaired glucose tolerance, and diabetes mellitus in the ELSA-Brasil study. CAD SAUDE PUBLICA 2021; 37:e00255920. [PMID: 34669776 DOI: 10.1590/0102-311x00255920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/22/2020] [Indexed: 11/21/2022] Open
Abstract
There is a conflict in the literature regarding the association between serum uric acid (SUA) levels and glycemic status. Therefore, we evaluated the association between SUA level and glycemic status - impaired fasting glucose (IFG), impaired glucose tolerance (IGT), and diabetes mellitus - and insulin resistance, in a large Brazilian study. This is a cross-sectional, observational study with 13,207 participants aged 35-74 years, at baseline (2008-2010) of the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil). A multinomial regression analysis was performed to test the association between SUA and glycemic status (IFG, IGT, and newly diagnosed type 2 diabetes at the cohort baseline) after adjustments by age, sex, skin color, body mass index, physical activity, smoking, alcohol consumption, comorbidities, and medicines use. Logistic regression model was used to evaluate the association between SUA and insulin resistance by HOMA-IR. Stratified analyses by sex were performed. The mean age (standard deviation) was 51.4 (8.9) years, 55.2% of participants were women. There were 1,439 newly diagnosed diabetes. After all adjustments, higher SUA was associated with IFG, IGT, and diabetes, with odds ratio (OR) = 1.15 (95%CI: 1.06; 1.25), 1.23 (95%CI: 1.14; 1.33), and 1.37 (95%CI: 1.24; 1.51), respectively. There was association between SUA levels and insulin resistance with OR = 1.24 (95%CI: 1.13; 1.36). In analysis stratified by sex, higher SUA persisted independently associated with impaired glycemic status. Our results suggest that a higher SUA levels were significantly associated with glycemic status in a large Latin American population, mainly among women.
Collapse
Affiliation(s)
| | | | | | | | - Maria Inês Schmidt
- Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brasil
| | | | - Sandhi Maria Barreto
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | | |
Collapse
|
40
|
Zhang W, Jin Y, Li J, Huang J, Chen H. Effects of genetic and nongenetic factors on hyperuricemia in Chinese patients with coronary artery disease. Pharmacogenomics 2021; 22:821-831. [PMID: 34505535 DOI: 10.2217/pgs-2021-0053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aim: The relationship between hyperuricemia and polymorphisms of transporter genes in coronary artery disease (CAD) patients in China remains unclear. Materials & methods: A total of 258 hyperuricemia patients with CAD and 242 control patients with CAD were recruited in this case-control study. Twenty-four SNPs in genes of ABCG2, PDZK1, URAT1, OAT4, GLUT9, ABCC4, NPT1 and NPT4 were genotyped using direct sequencing in all subjects. Results: The mutation of ABCG2 rs2231142 locus increases the risk of hyperuricemia, and there is a gene dose effect in the influence of mutant heterozygotes and homozygotes. rs3825017 in URAT1 and rs62293298 in GLUT9 were also confirmed to be associated with hyperuricemia. Conclusion: Age, weight, creatinine clearance rate, diuretics and SNPs on ABCG2, URAT1 and GLUT9 were all risk factors of hyperuricemia.
Collapse
Affiliation(s)
- Weixia Zhang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiwen Jin
- Department ofPharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Juan Li
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingjing Huang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hefeng Chen
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Bernabeu E, Canela-Xandri O, Rawlik K, Talenti A, Prendergast J, Tenesa A. Sex differences in genetic architecture in the UK Biobank. Nat Genet 2021; 53:1283-1289. [PMID: 34493869 DOI: 10.1038/s41588-021-00912-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/12/2021] [Indexed: 01/05/2023]
Abstract
Males and females present differences in complex traits and in the risk of a wide array of diseases. Genotype by sex (GxS) interactions are thought to account for some of these differences. However, the extent and basis of GxS are poorly understood. In the present study, we provide insights into both the scope and the mechanism of GxS across the genome of about 450,000 individuals of European ancestry and 530 complex traits in the UK Biobank. We found small yet widespread differences in genetic architecture across traits. We also found that, in some cases, sex-agnostic analyses may be missing trait-associated loci and looked into possible improvements in the prediction of high-level phenotypes. Finally, we studied the potential functional role of the differences observed through sex-biased gene expression and gene-level analyses. Our results suggest the need to consider sex-aware analyses for future studies to shed light onto possible sex-specific molecular mechanisms.
Collapse
Affiliation(s)
- Elena Bernabeu
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Oriol Canela-Xandri
- Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Konrad Rawlik
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Andrea Talenti
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - James Prendergast
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Albert Tenesa
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, UK.
- Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK.
| |
Collapse
|
42
|
Mandal AK, Leask MP, Estiverne C, Choi HK, Merriman TR, Mount DB. Genetic and Physiological Effects of Insulin on Human Urate Homeostasis. Front Physiol 2021; 12:713710. [PMID: 34408667 PMCID: PMC8366499 DOI: 10.3389/fphys.2021.713710] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/02/2021] [Indexed: 12/19/2022] Open
Abstract
Insulin and hyperinsulinemia reduce renal fractional excretion of urate (FeU) and play a key role in the genesis of hyperuricemia and gout, via uncharacterized mechanisms. To explore this association further we studied the effects of genetic variation in insulin-associated pathways on serum urate (SU) levels and the physiological effects of insulin on urate transporters. We found that urate-associated variants in the human insulin (INS), insulin receptor (INSR), and insulin receptor substrate-1 (IRS1) loci associate with the expression of the insulin-like growth factor 2, IRS1, INSR, and ZNF358 genes; additionally, we found genetic interaction between SLC2A9 and the three loci, most evident in women. We also found that insulin stimulates the expression of GLUT9 and increases [14C]-urate uptake in human proximal tubular cells (PTC-05) and HEK293T cells, transport activity that was effectively abrogated by uricosurics or inhibitors of protein tyrosine kinase (PTK), PI3 kinase, MEK/ERK, or p38 MAPK. Heterologous expression of individual urate transporters in Xenopus oocytes revealed that the [14C]-urate transport activities of GLUT9a, GLUT9b, OAT10, OAT3, OAT1, NPT1 and ABCG2 are directly activated by insulin signaling, through PI3 kinase (PI3K)/Akt, MEK/ERK and/or p38 MAPK. Given that the high-capacity urate transporter GLUT9a is the exclusive basolateral exit pathway for reabsorbed urate from the renal proximal tubule into the blood, that insulin stimulates both GLUT9 expression and urate transport activity more than other urate transporters, and that SLC2A9 shows genetic interaction with urate-associated insulin-signaling loci, we postulate that the anti-uricosuric effect of insulin is primarily due to the enhanced expression and activation of GLUT9.
Collapse
Affiliation(s)
- Asim K. Mandal
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Megan P. Leask
- Biochemistry Department, University of Otago, Dunedin, New Zealand
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, AL, United States
| | - Christopher Estiverne
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Hyon K. Choi
- Division of Rheumatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Tony R. Merriman
- Biochemistry Department, University of Otago, Dunedin, New Zealand
- Division of Rheumatology and Clinical Immunology, University of Alabama, Birmingham, AL, United States
| | - David B. Mount
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Renal Division, VA Boston Healthcare System, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Wang L, Balmat TJ, Antonia AL, Constantine FJ, Henao R, Burke TW, Ingham A, McClain MT, Tsalik EL, Ko ER, Ginsburg GS, DeLong MR, Shen X, Woods CW, Hauser ER, Ko DC. An atlas connecting shared genetic architecture of human diseases and molecular phenotypes provides insight into COVID-19 susceptibility. Genome Med 2021; 13:83. [PMID: 34001247 PMCID: PMC8127495 DOI: 10.1186/s13073-021-00904-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND While genome-wide associations studies (GWAS) have successfully elucidated the genetic architecture of complex human traits and diseases, understanding mechanisms that lead from genetic variation to pathophysiology remains an important challenge. Methods are needed to systematically bridge this crucial gap to facilitate experimental testing of hypotheses and translation to clinical utility. RESULTS Here, we leveraged cross-phenotype associations to identify traits with shared genetic architecture, using linkage disequilibrium (LD) information to accurately capture shared SNPs by proxy, and calculate significance of enrichment. This shared genetic architecture was examined across differing biological scales through incorporating data from catalogs of clinical, cellular, and molecular GWAS. We have created an interactive web database (interactive Cross-Phenotype Analysis of GWAS database (iCPAGdb)) to facilitate exploration and allow rapid analysis of user-uploaded GWAS summary statistics. This database revealed well-known relationships among phenotypes, as well as the generation of novel hypotheses to explain the pathophysiology of common diseases. Application of iCPAGdb to a recent GWAS of severe COVID-19 demonstrated unexpected overlap of GWAS signals between COVID-19 and human diseases, including with idiopathic pulmonary fibrosis driven by the DPP9 locus. Transcriptomics from peripheral blood of COVID-19 patients demonstrated that DPP9 was induced in SARS-CoV-2 compared to healthy controls or those with bacterial infection. Further investigation of cross-phenotype SNPs associated with both severe COVID-19 and other human traits demonstrated colocalization of the GWAS signal at the ABO locus with plasma protein levels of a reported receptor of SARS-CoV-2, CD209 (DC-SIGN). This finding points to a possible mechanism whereby glycosylation of CD209 by ABO may regulate COVID-19 disease severity. CONCLUSIONS Thus, connecting genetically related traits across phenotypic scales links human diseases to molecular and cellular measurements that can reveal mechanisms and lead to novel biomarkers and therapeutic approaches. The iCPAGdb web portal is accessible at http://cpag.oit.duke.edu and the software code at https://github.com/tbalmat/iCPAGdb .
Collapse
Affiliation(s)
- Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Thomas J Balmat
- Duke Research Computing, Duke University, Durham, NC, 27710, USA
| | - Alejandro L Antonia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Florica J Constantine
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Ricardo Henao
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Thomas W Burke
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Andy Ingham
- Duke Research Computing, Duke University, Durham, NC, 27710, USA
| | - Micah T McClain
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC, 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ephraim L Tsalik
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC, 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Emily R Ko
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Department of Hospital Medicine, Duke Regional Hospital, Durham, NC, 27705, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Mark R DeLong
- Duke Research Computing, Duke University, Durham, NC, 27710, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Woo Center for Big Data and Precision Health, Duke University, Durham, NC, 27710, USA
| | - Christopher W Woods
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC, 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Elizabeth R Hauser
- Duke Molecular Physiology Institute and Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, 27710, USA
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, NC, 27705, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA.
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
44
|
Laskar RS, Li P, Ecsedi S, Abedi-Ardekani B, Durand G, Robinot N, Hubert JN, Janout V, Zaridze D, Mukeria A, Mates D, Holcatova I, Foretova L, Swiatkowska B, Dzamic Z, Milosavljevic S, Olaso R, Boland A, Deleuze JF, Muller DC, McKay JD, Brennan P, Le Calvez-Kelm F, Scelo G, Chanudet E. Sexual dimorphism in cancer: insights from transcriptional signatures in kidney tissue and renal cell carcinoma. Hum Mol Genet 2021; 30:343-355. [PMID: 33527138 PMCID: PMC8098110 DOI: 10.1093/hmg/ddab031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
Sexual dimorphism in cancer incidence and outcome is widespread. Understanding the underlying mechanisms is fundamental to improve cancer prevention and clinical management. Sex disparities are particularly striking in kidney cancer: across diverse populations, men consistently show unexplained 2-fold increased incidence and worse prognosis. We have characterized genome-wide expression and regulatory networks of 609 renal tumors and 256 non-tumor renal tissues. Normal kidney displayed sex-specific transcriptional signatures, including higher expression of X-linked tumor suppressor genes in women. Sex-dependent genotype-phenotype associations unraveled women-specific immune regulation. Sex differences were markedly expanded in tumors, with male-biased expression of key genes implicated in metabolism, non-malignant diseases with male predominance and carcinogenesis, including markers of tumor infiltrating leukocytes. Analysis of sex-dependent RCC progression and survival uncovered prognostic markers involved in immune response and oxygen homeostasis. In summary, human kidney tissues display remarkable sexual dimorphism at the molecular level. Sex-specific transcriptional signatures further shape renal cancer, with relevance for clinical management.
Collapse
Affiliation(s)
- Ruhina S Laskar
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Peng Li
- Laboratory of Population Health, Max Planck Institute for Demographic Research, 18057 Rostock, Germany
| | - Szilvia Ecsedi
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Behnoush Abedi-Ardekani
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Geoffroy Durand
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Nivonirina Robinot
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Jean-Noël Hubert
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Vladimir Janout
- Science and Research Center, Faculty of Health Sciences, Palacky University, 77900 Olomouc, Czech Republic
| | - David Zaridze
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Centre, 115478 Moscow, Russian Federation
| | - Anush Mukeria
- Department of Epidemiology and Prevention, Russian N.N. Blokhin Cancer Research Centre, 115478 Moscow, Russian Federation
| | - Dana Mates
- Department of Environmental Health, National Institute of Public Health, 050463 Bucharest, Romania
| | - Ivana Holcatova
- Department of Public Health and Preventive Medicine, Charles University, Second Faculty of Medicine, 15006 Prague, Czech Republic
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, 60200 Brno, Czech Republic
| | - Beata Swiatkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, 91-348 Lodz, Poland
| | - Zoran Dzamic
- Clinic of Urology, Clinical Center of Serbia (KCS), University of Belgrade - Faculty of Medicine, 11000 Belgrade, Serbia
| | - Sasa Milosavljevic
- International Organisation for Cancer Prevention and Research, 11070 Belgrade, Serbia
| | - Robert Olaso
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine, 91057, Evry, France
| | - David C Muller
- Faculty of Medicine, School of Public Health, Imperial College London, W21NY London, UK
| | - James D McKay
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Paul Brennan
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Florence Le Calvez-Kelm
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Ghislaine Scelo
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
- Unit of Cancer Epidemiology, Department of Medical Sciences, University of Turin, 8-10124 Turin, Italy
| | - Estelle Chanudet
- Section of Genetics, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| |
Collapse
|
45
|
Mehmood A, Zhao L, Ishaq M, Xin W, Zhao L, Wang C, Hossen I, Zhang H, Lian Y, Xu M. Anti-hyperuricemic potential of stevia (Stevia rebaudiana Bertoni) residue extract in hyperuricemic mice. Food Funct 2021; 11:6387-6406. [PMID: 32613954 DOI: 10.1039/c9fo02246e] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Hyperuricemia (HUA) is considered a potent risk factor for the development of gout, renal failure, and cardiovascular disease. The current project was designed to use stevia (Stevia rebaudiana Bertoni) byproduct, named stevia residue extract (STVRE), for the treatment of HUA. Male Kunming mice were divided into six groups: normal control, model control, positive control (allopurinol, 5 mg per kg body weight [bw]), STVRE-1 (75 mg per kg bw), STVRE-2 (150 mg per kg bw), and STVRE-3 (300 mg per kg bw). HUA was induced by the administration of potassium oxonate (100 mg per kg bw), fructose (10% w/v), and yeast extract (100 mg per kg bw) for 8 weeks. STVRE significantly (p < 0.05) decreased uric acid (UA) production and ameliorated UA excretion by interacting with urate transporters. The STVRE remarkably attenuated oxidative stress mediated by UA and downregulated inflammatory-related response markers such as COX-2, NF-κB, PGE2, IL-1β, and TNF-α. Furthermore, STVRE also reversed HUA-induced abnormalities in kidneys compared with the MC group. The results of our study suggest that STVRE has potential to attenuate hyperuricemia and renal protective effects, and may be used as a natural supplement for the possible treatment of UA-related disorders.
Collapse
Affiliation(s)
- Arshad Mehmood
- Beijing Advance Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives School of Food and Chemical Technology, Beijing Technology and Business University, Beijing 100048, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Halbritter J. Genetics of kidney stone disease-Polygenic meets monogenic. Nephrol Ther 2021; 17S:S88-S94. [PMID: 33910705 DOI: 10.1016/j.nephro.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 11/16/2022]
Abstract
Kidney stone disease comprising nephrolithiasis and nephrocalcinosis is a clinical syndrome of increasing prevalence with remarkable heterogeneity. Stone composition, age of manifestation, rate of recurrence, and impairment of kidney function varies with underlying etiologies. While calcium-based kidney stones account for the vast majority their etiology is still poorly understood. Recent studies underline the notion that genetic susceptibility together with dietary habits constitutes the major driver of kidney stone formation. In addition to single gene (Mendelian) disorders, which are most likely underestimated in the adult population, common risk alleles explain part of the observed heritability. Interestingly, identified GWAS loci often match those of Mendelian disease genes and vice versa (CASR, SLC34A1, CYP24A1). These findings provide mechanistic links related to renal calcium homeostasis, vitamin D metabolism, and CaSR-signaling regulated by the CaSR-CLDN14-CLDN16/19 axis (paracellular Ca2+ reabsorption) and TRPV5 (transcellular Ca2+ reabsorption). Recent identification of new single gene disorders of calcium-oxalate-nephrolithiasis (SLC26A1, CLDN2) and distal renal tubular acidosis with nephrocalcinosis (FOXI1, WDR72, ATP6V1C2) enabled additional insights into the kidney-gut axis and molecular prerequisites of proper urinary acidification. Implementation of centralized patient registries on hereditary kidney stone diseases are necessary to build up well characterized cohorts for urgently needed clinical studies.
Collapse
Affiliation(s)
- Jan Halbritter
- Medical Department III, Endocrinology, Nephrology and Rheumatology, Division of Nephrology, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
47
|
Mozzini C, Girelli D, Setti A, Croce J, Stefanoni F, Castagna A, Pizzolo F, Friso S, Olivieri O, Martinelli N. Serum Uric Acid Levels, but Not rs7442295 Polymorphism of SCL2A9 Gene, Predict Mortality in Clinically Stable Coronary Artery Disease. Curr Probl Cardiol 2021; 46:100798. [PMID: 33540324 DOI: 10.1016/j.cpcardiol.2021.100798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 10/22/2022]
Abstract
Serum uric acid (SUA) has been associated with cardiovascular disease, but up to now whether SUA is an independent cardiovascular risk factor or merely a disease-related epiphenomenon remains still controversial. within the framework of the Verona Heart Study, we prospectively followed 703 subjects with angiographically demonstrated and clinically stable coronary artery disease between May 1996 and March 2007. At baseline, SUA levels were measured in all the patients. Genotype data of SCL2A9 rs7442295 polymorphism, which has been associated with SUA by genome-wide association studies, were available for 686 subjects (97.6%). After a median follow-up of 57 months, 116 patients (16.5%) had died, 83 (11.8%) because of cardiovascular causes. Patients with hyperuricemia, defined by SUA levels above the 75th percentile (≥0.41 mmol/L), had an increased total and cardiovascular mortality rate than those with SUA below this threshold level (23.3% vs 14.1%, P = 0.048 and 19.4% vs 9.2%, P = 0.001, respectively, by Kaplan-Meier with Log-Rank test). These associations were confirmed by Cox regression after adjustment for sex, age, other predictors of mortality, coronary revascularization, and drug therapies at discharge (hazard ratio for total mortality 1.87 [1.05-3.34], P = 0.033; hazard ratio for cardiovascular mortality 2.09 [1.03-4.25], P = 0.041). Although associated with SUA levels, rs7442295 polymorphism did not predict total or cardiovascular mortality. our data support that SUA may be a prognostic cardiovascular biomarker, predicting total and cardiovascular mortality in the setting of secondary prevention of coronary artery disease. On the other hand, SCL2A9 gene polymorphism, notwithstanding a clear influence on SUA levels, was not associated with mortality.
Collapse
Affiliation(s)
- Chiara Mozzini
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy.
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Angela Setti
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Jacopo Croce
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Filippo Stefanoni
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Francesca Pizzolo
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Simonetta Friso
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Oliviero Olivieri
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| | - Nicola Martinelli
- Department of Medicine, Section of Internal Medicine, University of Verona, Verona, Italy
| |
Collapse
|
48
|
Wang L, Balmat TJ, Antonia AL, Constantine FJ, Henao R, Burke TW, Ingham A, McClain MT, Tsalik EL, Ko ER, Ginsburg GS, DeLong MR, Shen X, Woods CW, Hauser ER, Ko DC. An atlas connecting shared genetic architecture of human diseases and molecular phenotypes provides insight into COVID-19 susceptibility. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.12.20.20248572. [PMID: 33398303 PMCID: PMC7781346 DOI: 10.1101/2020.12.20.20248572] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
While genome-wide associations studies (GWAS) have successfully elucidated the genetic architecture of complex human traits and diseases, understanding mechanisms that lead from genetic variation to pathophysiology remains an important challenge. Methods are needed to systematically bridge this crucial gap to facilitate experimental testing of hypotheses and translation to clinical utility. Here, we leveraged cross-phenotype associations to identify traits with shared genetic architecture, using linkage disequilibrium (LD) information to accurately capture shared SNPs by proxy, and calculate significance of enrichment. This shared genetic architecture was examined across differing biological scales through incorporating data from catalogs of clinical, cellular, and molecular GWAS. We have created an interactive web database (interactive Cross-Phenotype Analysis of GWAS database (iCPAGdb); http://cpag.oit.duke.edu) to facilitate exploration and allow rapid analysis of user-uploaded GWAS summary statistics. This database revealed well-known relationships among phenotypes, as well as the generation of novel hypotheses to explain the pathophysiology of common diseases. Application of iCPAGdb to a recent GWAS of severe COVID-19 demonstrated unexpected overlap of GWAS signals between COVID-19 and human diseases, including with idiopathic pulmonary fibrosis driven by the DPP9 locus. Transcriptomics from peripheral blood of COVID-19 patients demonstrated that DPP9 was induced in SARS-CoV-2 compared to healthy controls or those with bacterial infection. Further investigation of cross-phenotype SNPs with severe COVID-19 demonstrated colocalization of the GWAS signal of the ABO locus with plasma protein levels of a reported receptor of SARS-CoV-2, CD209 (DC-SIGN), pointing to a possible mechanism whereby glycosylation of CD209 by ABO may regulate COVID-19 disease severity. Thus, connecting genetically related traits across phenotypic scales links human diseases to molecular and cellular measurements that can reveal mechanisms and lead to novel biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | | | - Alejandro L. Antonia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Florica J. Constantine
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Ricardo Henao
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Thomas W. Burke
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Andy Ingham
- Duke Research Computing, Duke University, Durham, NC 27710, USA
| | - Micah T. McClain
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Ephraim L. Tsalik
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Emily R. Ko
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Department of Hospital Medicine, Duke Regional Hospital, Durham, NC, 27705, USA
| | - Geoffrey S. Ginsburg
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Mark R. DeLong
- Duke Research Computing, Duke University, Durham, NC 27710, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Woo Center for Big Data and Precision Health, Duke University, Durham, NC 27710, USA
| | - Christopher W. Woods
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Elizabeth R. Hauser
- Duke Molecular Physiology Institute and Department of Biostatistics and Bioinformatics, Duke University Medical Center Durham, NC 27710, USA
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, NC 27705, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Lead contact
| |
Collapse
|
49
|
Abstract
Uric acid, the end product of purine metabolism, plays a key role in the pathogenesis of gout and other disease processes. The circulating serum uric acid concentration is governed by the relative balance of hepatic production, intestinal secretion, and renal tubular reabsorption and secretion. An elegant synergy between genome-wide association studies and transport physiology has led to the identification and characterization of the major transporters involved with urate reabsorption and secretion, in both kidney and intestine. This development, combined with continued analysis of population-level genetic data, has yielded an increasingly refined mechanistic understanding of uric acid homeostasis as well as greater understanding of the genetic and acquired influences on serum uric acid concentration. The continued delineation of novel and established regulatory pathways that regulate uric acid homeostasis promises to lead to a more complete understanding of uric acid-associated diseases and to identify new targets for treatment.
Collapse
Affiliation(s)
| | - Asim K Mandal
- Renal Division, Brigham and Women's Hospital, Boston, MA
| | - David B Mount
- Renal Division, Brigham and Women's Hospital, Boston, MA; Renal Division, VA Boston Healthcare System, Harvard Medical School, Boston, MA.
| |
Collapse
|
50
|
Lukkunaprasit T, Rattanasiri S, Turongkaravee S, Suvannang N, Ingsathit A, Attia J, Thakkinstian A. The association between genetic polymorphisms in ABCG2 and SLC2A9 and urate: an updated systematic review and meta-analysis. BMC MEDICAL GENETICS 2020; 21:210. [PMID: 33087043 PMCID: PMC7580000 DOI: 10.1186/s12881-020-01147-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/13/2020] [Indexed: 02/08/2023]
Abstract
Background Replication studies showed conflicting effects of ABCG2 and SLC2A9 polymorphisms on gout and serum urate. This meta-analysis therefore aimed to pool their effects across studies. Methods Studies were located from MEDLINE and Scopus from inception to 17th June 2018. Observational studies in adults with any polymorphism in ABCG2 or SLC2A9, and outcome including gout, hyperuricemia, and serum urate were included for pooling. Data extractions were performed by two independent reviewers. Genotype effects were pooled stratified by ethnicity using a mixed-effect logistic model and a multivariate meta-analysis for dichotomous and continuous outcomes. Results Fifty-two studies were included in the analysis. For ABCG2 polymorphisms, mainly studied in Asians, carrying 1–2 minor-allele-genotypes of rs2231142 and rs72552713 were respectively about 2.1–4.5 and 2.5–3.9 times higher odds of gout than non-minor-allele-genotypes. The two rs2231142-risk-genotypes also had higher serum urate about 11–18 μmol/l. Conversely, carrying 1–2 minor alleles of rs2231137 was about 36–57% significantly lower odds of gout. For SLC2A9 polymorphisms, mainly studied in Caucasians, carrying 1–2 minor alleles of rs1014290, rs6449213, rs6855911, and rs7442295 were about 25–43%, 31–62%, 33–64%, and 35–65% significantly lower odds of gout than non-minor-allele-genotypes. In addition, 1–2 minor-allele-genotypes of the latter three polymorphisms had significantly lower serum urate about 20–49, 21–51, and 18–54 μmol/l than non-minor-allele-genotypes. Conclusions Our findings should be useful in identifying patients at risk for gout and high serum urate and these polymorphisms may be useful in personalized risk scores. Trial registration PROSPERO registration number: CRD42018105275. Supplementary information The online version contains supplementary material available at 10.1186/s12881-020-01147-2.
Collapse
Affiliation(s)
- Thitiya Lukkunaprasit
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd., Ratchathewi, Bangkok, 10400, Thailand.,Department of Pharmacology, College of Pharmacy, Rangsit University, Pathum Thani, Thailand
| | - Sasivimol Rattanasiri
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd., Ratchathewi, Bangkok, 10400, Thailand.
| | - Saowalak Turongkaravee
- Social and Administrative Pharmacy Excellence Research (SAPER) Unit, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Naravut Suvannang
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd., Ratchathewi, Bangkok, 10400, Thailand
| | - Atiporn Ingsathit
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd., Ratchathewi, Bangkok, 10400, Thailand
| | - John Attia
- Centre for Clincial Epidemiology and Biostatistics, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ammarin Thakkinstian
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd., Ratchathewi, Bangkok, 10400, Thailand
| |
Collapse
|