1
|
Takase M, Nakaya N, Nakamura T, Kogure M, Hatanaka R, Nakaya K, Chiba I, Tokioka S, Kanno I, Nochioka K, Tsuchiya N, Hirata T, Narita A, Obara T, Ishikuro M, Ohseto H, Uruno A, Kobayashi T, Kodama EN, Hamanaka Y, Orui M, Ogishima S, Nagaie S, Fuse N, Sugawara J, Kuriyama S. Genetic risk, lifestyle adherence, and risk of developing hyperuricaemia in a Japanese population. Rheumatology (Oxford) 2025; 64:2591-2600. [PMID: 39271169 PMCID: PMC12048061 DOI: 10.1093/rheumatology/keae492] [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: 04/26/2024] [Revised: 07/16/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
OBJECTIVE The objective of this study was to investigate the inter-relationships among genetic risk, adherence to a healthy lifestyle, and susceptibility to hyperuricaemia. METHODS This prospective cohort study was conducted with 7241 hyperuricaemia-free individuals aged ≥20 years from the Tohoku Medical Megabank Community-based cohort study. A comprehensive lifestyle score included assessment of BMI, smoking, drinking, and physical activity, and a polygenic risk score (PRS) was constructed based on uric acid loci from a previous genome-wide association study meta-analysis. A multiple logistic regression model was used to estimate the association between genetic risk, adherence to a healthy lifestyle, and hyperuricaemia incidence and to calculate the area under the receiver operating characteristic curve (AUROC). Hyperuricaemia was defined as a uric acid level of ≥7.0 mg/dL or a self-reported history of hyperuricaemia. RESULTS Of the 7241 adults [80.7% females; mean (±s.d.) age: 57.7 (12.6) years], 217 (3.0%) developed hyperuricaemia during 3.5 years of follow-up period. Genetic risk was correlated with hyperuricaemia development (P for interaction = 0.287), and lifestyle risks were independently associated. Participants with a high genetic risk and poor lifestyle had the highest risk (odds ratio: 5.34; 95% CI: 2.61-12.10). Although not statistically significant, adding the PRS in the model with lifestyle information improved predictive ability (AUROC = 0.771, 95% CI: 0.736-0.806 for lifestyle; AUROC = 0.785, 95% CI: 0.751-0.819 for lifestyle and PRS; P= 0.07). CONCLUSION A healthy lifestyle to prevent hyperuricaemia, irrespective of genetic risk, may mitigate the genetic risk. Genetic risk may complement lifestyle factors in identifying individuals at a heightened hyperuricaemia risk.
Collapse
Affiliation(s)
- Masato Takase
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Naoki Nakaya
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Tomohiro Nakamura
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Kyoto Women’s University, Kyoto, Japan
| | - Mana Kogure
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Rieko Hatanaka
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Kumi Nakaya
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Ippei Chiba
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Sayuri Tokioka
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Ikumi Kanno
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Kotaro Nochioka
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Sendai, Miyagi, Japan
| | - Naho Tsuchiya
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Takumi Hirata
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Human Care Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Tokyo, Japan
| | - Akira Narita
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Taku Obara
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Mami Ishikuro
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Hisashi Ohseto
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Akira Uruno
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Tomoko Kobayashi
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Sendai, Miyagi, Japan
| | - Eiichi N Kodama
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Miyagi, Japan
| | - Yohei Hamanaka
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Masatsugu Orui
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Soichi Ogishima
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Satoshi Nagaie
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Nobuo Fuse
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Junichi Sugawara
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Hospital, Tohoku University, Sendai, Miyagi, Japan
- Suzuki Memorial Hospital, Iwanumashi, Miyagi, Japan
| | - Shinichi Kuriyama
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
2
|
Hu S, Dalbeth N, Li Z, Chen Y, Dong B, Wang C, Sun W, Li C, Lu J. Associations of the ALDH2 rs671 polymorphism and alcohol intake with early-onset gout in a Chinese male cohort. Joint Bone Spine 2025; 92:105876. [PMID: 40015360 DOI: 10.1016/j.jbspin.2025.105876] [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: 10/15/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE Gout in young people is increasingly common across the world, including in China. This study aimed to identify clinical and genetic associations with early-onset gout in Chinese men. METHODS One thousand two hundred and one Chinese men with gout were included. Early-onset gout was defined as the first presentation of gout at <30years. Twenty single nucleotide polymorphisms (SNPs) identified as gout-risk loci or associated with serum urate (SU) levels in East Asian populations were genotyped. Logistic regression was used to evaluate the association of SNPs and clinical factors with early-onset gout. RESULTS Four hundred and thirty-three (36.1%) participants were identified as having early-onset gout. These patients had higher SU levels and were more likely to experience gout flares than those with later-onset gout. The ALDH2 rs671 GG genotype was associated with a lower risk of early-onset gout. Compared to those with GG genotype who never drank alcohol before gout onset, individuals with AA or AG genotypes who drank alcohol before gout onset had a higher likelihood of early-onset gout. Additionally, alcohol intake significantly increased the likelihood of gout flares in early-onset gout patients. Moreover, body mass index, sugar-sweetened beverage intake, family history of gout and renal urate underexcretion were associated with early-onset gout. CONCLUSIONS The ALDH2 rs671 GG genotype was significantly associated with a lower risk of early-onset gout, while individuals with the AA or AG genotype who consumed alcohol were more susceptible. These findings indicate that alcohol intake is a potentially modifiable risk factor for early-onset gout in genetically susceptible individuals.
Collapse
Affiliation(s)
- Shuhui Hu
- Shandong Provincial Clinical Research Center for Immune Diseases, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, 1023 Auckland, New Zealand
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, 266071 Qingdao, China
| | - Ying Chen
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China
| | - Bingzi Dong
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China
| | - Can Wang
- Shandong Provincial Clinical Research Center for Immune Diseases, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China
| | - Wenyan Sun
- Shandong Provincial Clinical Research Center for Immune Diseases, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China
| | - Changgui Li
- Shandong Provincial Clinical Research Center for Immune Diseases, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China
| | - Jie Lu
- Shandong Provincial Clinical Research Center for Immune Diseases, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, 266003 Qingdao, China.
| |
Collapse
|
3
|
Kalwick M, Roth M. A Comprehensive Review of the Genetics of Dyslipidemias and Risk of Atherosclerotic Cardiovascular Disease. Nutrients 2025; 17:659. [PMID: 40004987 PMCID: PMC11858766 DOI: 10.3390/nu17040659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Dyslipidemias are often diagnosed based on an individual's lipid panel that may or may not include Lp(a) or apoB. But these values alone omit key information that can underestimate risk and misdiagnose disease, which leads to imprecise medical therapies that reduce efficacy with unnecessary adverse events. For example, knowing whether an individual's dyslipidemia is monogenic can granularly inform risk and create opportunities for precision therapeutics. This review explores the canonical and non-canonical causes of dyslipidemias and how they impact atherosclerotic cardiovascular disease (ASCVD) risk. This review emphasizes the multitude of genetic causes that cause primary hypercholesterolemia, hypertriglyceridemia, and low or elevated high-density lipoprotein (HDL)-cholesterol levels. Within each of these sections, this review will explore the evidence linking these genetic conditions with ASCVD risk. Where applicable, this review will summarize approved therapies for a particular genetic condition.
Collapse
Affiliation(s)
| | - Mendel Roth
- GBinsight, GB Healthwatch, San Diego, CA 92122, USA;
| |
Collapse
|
4
|
Li G, Du S, Yan S, Wang Y, Bu R, Cheng M, Zhang Y, Chen Q, Wu Y, Zhang X, Wang D, Wang T. Mechanism of Biqi capsules in the treatment of gout based on network pharmacology and experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118817. [PMID: 39284427 DOI: 10.1016/j.jep.2024.118817] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 11/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gout is a crystal-related arthropathy caused by monosodium urate (MSU) deposition, resulting from purine metabolism disorders and hyperuricemia (HUA). Gout belongs to the traditional medicine category of Bi syndrome. Biqi capsules (BQ) is a traditional Chinese medicine formula used to treat Bi syndrome. The BQ prescription is derived from the ancient prescription of Hua Tuo, a famous physician in the Han Dynasty. AIM OF THE STUDY To study the effect and mechanism of BQ in treating acute gouty arthritis (AGA) and HUA. MATERIALS AND METHODS Analyzing BQ's signaling pathways for gout treatment via network pharmacology. The HUA model was induced orally with adenine and potassium oxonate. The rat AGA model was established by MSU injection. In vitro, MH7A and RAW 246.7 cells were treated with LPS and MSU. Serum uric acid, creatinine, and urea nitrogen levels were evaluated. Kidney and ankle joint pathology was observed via HE staining. Inflammatory signaling pathway proteins, epithelial-mesenchymal transition (EMT) pathway proteins, and uric acid metabolism-related proteins were detected by Western blot. RESULTS 1780 potential targets for gout treatment were identified, and 1039 target proteins corresponding to BQ's active ingredients were obtained. Pathway enrichment analysis revealed BQ improved gout mainly through inflammatory pathways. Experimental results showed BQ could reduce serum uric acid level and increase uric acid clearance rate by regulating the expression of adenosine deaminase (ADA), and organic anion transporter 1 (OAT1) and glucose transporter 9 (GLUT9) in HUA mice. BQ could improve renal function and injury by inhibiting the NLRP3 pathway in HUA mice' kidneys. Additionally, BQ could alleviate ankle joint swelling and synovial injury, inhibit the TLR4/NLRP3 pathway, and reduce levels of inflammatory factors including interleukin 6 (IL-6), interleukin 1β (IL-1β), and tumor necrosis factor-alpha (TNF-α) in AGA rats. The main component of BQ, brucine, could inhibit the activation of NLRP3/NF-κB pathway induced by MSU and reduce the expression level of inflammatory factors (IL-6, IL-1β, and TNF-α) in macrophages. Brucine could inhibit the activation of the EMT pathway and reduce the expression level of inflammatory factors (IL-6, TNF-α) in human fibroblast-like synoviocytes (MH7A cells) induced by MSU. CONCLUSIONS BQ effectively reduced serum uric acid levels, improved kidney and joint damage, and ameliorated the inflammatory response caused by MSU. Its main component, brucine, effectively improved the inflammatory response and reduced the invasive ability of synoviocytes induced by MSU.
Collapse
Affiliation(s)
- Ge Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Simiao Du
- Tianjin Darentang Jingwanhong Pharmaceutical Co., Ltd., 20 Daming Road, Xiqing District, Tianjin, 300112, China
| | - Siya Yan
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Yang Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Ruizhen Bu
- Tianjin Darentang Jingwanhong Pharmaceutical Co., Ltd., 20 Daming Road, Xiqing District, Tianjin, 300112, China
| | - Meifang Cheng
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Yi Zhang
- Haihe Laboratory of Modern Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Qian Chen
- Haihe Laboratory of Modern Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Yuzheng Wu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China
| | - Xiangqi Zhang
- Tianjin Darentang Jingwanhong Pharmaceutical Co., Ltd., 20 Daming Road, Xiqing District, Tianjin, 300112, China.
| | - Dan Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Jinghai District, Tianjin, 301617, China.
| | - Tao Wang
- Haihe Laboratory of Modern Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
5
|
Biswas S, Hilser JR, Woodward NC, Wang Z, Gukasyan J, Nemet I, Schwartzman WS, Huang P, Han Y, Fouladian Z, Charugundla S, Spencer NJ, Pan C, Tang WHW, Lusis AJ, Hazen SL, Hartiala JA, Allayee H. Exploring the Role of Glycine Metabolism in Coronary Artery Disease: Insights from Human Genetics and Mouse Models. Nutrients 2025; 17:198. [PMID: 39796632 PMCID: PMC11723402 DOI: 10.3390/nu17010198] [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: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Circulating glycine levels have been associated with reduced risk of coronary artery disease (CAD) in humans but these associations have not been observed in all studies. We evaluated whether the relationship between glycine levels and atherosclerosis was causal using genetic analyses in humans and feeding studies in mice. Methods: Serum glycine levels were evaluated for association with risk of CAD in the UK Biobank. Genetic determinants of glycine levels were identified through a genome-wide association study (GWAS) and used to evaluate the causal relationship between glycine and risk of CAD by Mendelian randomization (MR). A dietary supplementation study was carried out with atherosclerosis-prone apolipoprotein E deficient (ApoE-/-) mice to determine the effects of increased circulating glycine levels on cardiometabolic traits and aortic lesion formation. Results: Among 105,718 UK Biobank subjects, elevated serum glycine levels were associated with significantly reduced risk of prevalent CAD (Quintile 5 vs. Quintile 1 OR = 0.76, 95% CI 0.67-0.87; p < 0.0001) and incident CAD (Quintile 5 vs. Quintile 1 HR = 0.70, 95% CI 0.65-0.77; p < 0.0001) after adjustment for age, sex, ethnicity, anti-hypertensive and lipid-lowering medications, blood pressure, kidney function, and diabetes. A GWAS meta-analysis with 230,947 subjects identified 61 loci for glycine levels, of which 26 were novel. MR analyses provided modest evidence that genetically elevated glycine levels were causally associated with reduced systolic blood pressure and risk of type 2 diabetes, but did not provide significant evidence for an association with decreased risk of CAD. Glycine supplementation in mice had no effects on cardiometabolic traits or atherosclerotic lesion development. Conclusions: While expanding the genetic architecture of glycine metabolism, MR analyses and in vivo feeding studies did not provide evidence that the clinical association of this amino acid with atherosclerosis represents a causal relationship.
Collapse
Affiliation(s)
- Subarna Biswas
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - James R. Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Nicholas C. Woodward
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Janet Gukasyan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ina Nemet
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
| | - William S. Schwartzman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pin Huang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yi Han
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zachary Fouladian
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Sarada Charugundla
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Neal J. Spencer
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Calvin Pan
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - W. H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Aldons J. Lusis
- Department of Medicine, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095, USA
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jaana A. Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
6
|
Asghari KM, Zahmatyar M, Seyedi F, Motamedi A, Zolfi M, Alamdary SJ, Fazlollahi A, Shamekh A, Mousavi SE, Nejadghaderi SA, Mohammadinasab R, Ghazi-Sha'rbaf J, Karamzad N, Sullman MJM, Kolahi AA, Safiri S. Gout: global epidemiology, risk factors, comorbidities and complications: a narrative review. BMC Musculoskelet Disord 2024; 25:1047. [PMID: 39702222 DOI: 10.1186/s12891-024-08180-9] [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: 04/11/2023] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Gout is one of the oldest known diseases and the most common form of inflammatory arthritis. The established risk factors for gout include hyperuricemia, chronic renal disease, genetic, alcohol consumption, dietary factors, diuretic use, hypertension, obesity, and metabolic syndrome. Patients with gout have an increased risk of all-cause mortality, particularly from cardiovascular disease, cancer, and infectious diseases. Gout is also associated with several complications, such as nephrolithiasis. This literature review describes the global epidemiology and trends associated with gout, before providing an overview of its risk factors and complications. METHODS This research used the narrative review method. Thorough searches were performed in PubMed and Google scholar, up to June 15, 2024, for articles that evaluated the risk factors, comorbidities or complications associated with gout. Moreover, we also included studies that reported the epidemiological characteristics or burden of gout at the global, regional, or national level. RESULTS Gout is more prevalent in developed countries, than in developing countries, although its prevalence is increasing globally. In addition, gout is much more prevalent among males than among females. Hyperuricemia has the largest role in the development of gout, although many risk factors contribute to the increasing prevalence of gout, including genes, several medications, and diet. Gout is associated with several comorbidities and complications, which need to be taken into consideration when managing gout. In recent years, gout has been found to be associated with several new comorbidities. CONCLUSIONS Our findings provide a comprehensive and informative overview that can be useful for the prevention, diagnosis, and management of gout.
Collapse
Affiliation(s)
- Kimia Motlagh Asghari
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Zahmatyar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Seyedi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Motamedi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maedeh Zolfi
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Asra Fazlollahi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Ehsan Mousavi
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Aria Nejadghaderi
- HIV/STI Surveillance Research Center, and WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ghazi-Sha'rbaf
- Department of Islamic History and Civilization, Faculty of Theology, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Nahid Karamzad
- Department of Persian Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark J M Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Safiri
- Neurosciences Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Flanagan J, Liu X, Ortega-Reyes D, Tomizuka K, Matoba N, Akiyama M, Koido M, Ishigaki K, Ashikawa K, Takata S, Shi M, Aoi T, Momozawa Y, Ito K, Murakami Y, Matsuda K, Kamatani Y, Morris AP, Horikoshi M, Terao C. Population-specific reference panel improves imputation quality for genome-wide association studies conducted on the Japanese population. Commun Biol 2024; 7:1665. [PMID: 39702642 DOI: 10.1038/s42003-024-07338-4] [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: 07/22/2023] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
To improve imputation quality for genome-wide association studies (GWAS) conducted on the Japanese population, we developed and evaluated four Japanese population-specific reference panels. These panels were constructed through the augmentation of the 1000 Genomes Project (1KG) panel using Japanese whole genome sequencing (WGS) data, with sample sizes ranging from 1 K to 7 K individuals enrolled through the Biobank Japan (BBJ) project, and sequencing depths ranging from 3× to 30×. Among these panels, an augmented reference panel comprising 7472 WGS samples of mixed depth (1KG+7K) exhibit the greatest improvement in imputation quality relative to the Trans-Omics for Precision Medicine (TOPMed) reference panel. Notably, we observe these improvements primarily for rare variants with a minor allele frequency (MAF) <5%. To demonstrate the benefits of improved imputation quality in association analyses of complex traits, we conducted GWAS for serum uric acid and total cholesterol levels following imputation up to the 1KG+7K panel. The analysis reveals several loci reaching genome-wide significance (P < 5 × 10-8) in the 1KG+7K imputation output yet remaining undetected when the same sample set is imputed up to the TOPMed reference panel. In summary, the 1KG+7K panel demonstrates significant advantages in the discovery of trait-associated loci, particularly those influenced by low-frequency association signals.
Collapse
Affiliation(s)
- Jack Flanagan
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Graduate School of Data Science, Seoul National University, Seoul, Republic of Korea
| | - Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - David Ortega-Reyes
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for DNA Data Analysis, National Institute of Genetics, Shizuoka, Japan
- Department of Genetics, School of Life Science, The Graduate University for Advanced Studies, SOKENDAI, Kanagawa, Japan
| | - Kohei Tomizuka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nana Matoba
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Genetics, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Masato Akiyama
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Koido
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kyota Ashikawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Sadaaki Takata
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - MingYang Shi
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomomi Aoi
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshinori Murakami
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, UK
| | - Momoko Horikoshi
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan.
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
8
|
He H, Cheng Q, Chen S, Li Q, Zhang J, Shan G, Zhang M. Triglyceride-glucose index and its additive interaction with ABCG2/SLC2A9 polygenic risk score on hyperuricemia in middle age and older adults: findings from the DLCC and BHMC study. Ann Med 2024; 56:2434186. [PMID: 39607832 DOI: 10.1080/07853890.2024.2434186] [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: 02/22/2024] [Revised: 05/03/2024] [Accepted: 09/26/2024] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVE We aim to investigate the joint effect of triglyceride-glucose (TyG) index and polygenic risk scores (PRS) of urate transporter genes ABCG2 and SLC2A9 on hyperuricemia. METHODS Baseline data from two prospective population-based cohort studies, including 30,453 individuals aged 50 years or older, were used to analyze the association between TyG index and hyperuricemia. A case-control study was then designed from the cohorts to investigate the interaction between genetic predisposition and TyG index on hyperuricemia among 595 matched pairs. PRS was constructed using 14 single nucleotide polymorphisms located in the ABCG2 and SLCA29 genes. RESULTS In both sexes, higher TyG index levels were correlated with elevated serum urate (SUA) levels (p values in both sexes < 0.001). In men, per unit increase of TyG was associated with a 1.44-fold (95% confidence interval [CI]: 1.35-1.55) higher risk of hyperuricemia after adjusted for covariates. In women, this estimate was 1.69 (1.51-1.89). Demonstrated by the restrict cubic spline model, TyG index was both linearly and non-linearly associated with elevated SUA (both p values < 0.001). Association between TyG index and hyperuricemia was stronger among people with higher genetic risk, and vice versa. Compared to people with TyG < 9 and PRS < 2, the odds ratios (ORs) (95% CIs) for hyperuricemia in the TyG <9 but PRS ≥2, TyG ≥9 but PRS < 2, TyG ≥9 and PRS ≥2 groups were 3.30 (1.53-7.14), 3.16 (1.23-8.11) and 7.55 (2.76-20.65), respectively. Additive interaction was also significant, with 57.5% (30.5%-84.4%) of the excess risk attributable to the additive gene-TyG index interaction. CONCLUSIONS The impact of genetic predisposition on hyperuricemia was significantly greater among individuals with a higher TyG index. Over 50% of the increased risk can be attributed to the interaction, indicating a crucial synergy between genetic factors and TyG index when estimating hyperuricemia risk.
Collapse
Affiliation(s)
- Huijing He
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Qiaolu Cheng
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Shuo Chen
- Beijing Physical Examination Center, Beijing, China
| | - Qiang Li
- Beijing Physical Examination Center, Beijing, China
| | - Jingbo Zhang
- Integrated Office, Beijing Medical Science and Technology Promotion Center, China
| | - Guangliang Shan
- Department of Epidemiology and Statistics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| | - Minying Zhang
- School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
9
|
Shine BK, Choi JE, Park YJ, Hong KW. The Genetic Variants Influencing Hypertension Prevalence Based on the Risk of Insulin Resistance as Assessed Using the Metabolic Score for Insulin Resistance (METS-IR). Int J Mol Sci 2024; 25:12690. [PMID: 39684400 DOI: 10.3390/ijms252312690] [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: 10/21/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Insulin resistance is a major indicator of cardiovascular diseases, including hypertension. The Metabolic Score for Insulin Resistance (METS-IR) offers a simplified and cost-effective way to evaluate insulin resistance. This study aimed to identify genetic variants associated with the prevalence of hypertension stratified by METS-IR score levels. Data from the Korean Genome and Epidemiology Study (KoGES) were analyzed. The METS-IR was calculated using the following formula: ln [(2 × fasting blood glucose (FBG) + triglycerides (TG)) × body mass index (BMI)]/ ln [high-density lipoprotein cholesterol (HDL-C)]. The participants were divided into tertiles 1 (T1) and 3 (T3) based on their METS-IR scores. Genome-wide association studies (GWAS) were performed for hypertensive cases and non-hypertensive controls within these tertile groups using logistic regression adjusted for age, sex, and lifestyle factors. Among the METS-IR tertile groups, 3517 of the 19,774 participants (17.8%) at T1 had hypertension, whereas 8653 of the 20,374 participants (42.5%) at T3 had hypertension. A total of 113 single-nucleotide polymorphisms (SNPs) reached the GWAS significance threshold (p < 5 × 10-8) in at least one tertile group, mapping to six distinct genetic loci. Notably, four loci, rs11899121 (chr2p24), rs7556898 (chr2q24.3), rs17249754 (ATP2B1), and rs1980854 (chr20p12.2), were significantly associated with hypertension in the high-METS-score group (T3). rs10857147 (FGF5) was significant in both the T1 and T3 groups, whereas rs671 (ALDH2) was significant only in the T1 group. The GWASs identified six genetic loci significantly associated with hypertension, with distinct patterns across METS-IR tertiles, highlighting the role of metabolic context in genetic susceptibility. These findings underscore critical genetic factors influencing hypertension prevalence and provide insights into the metabolic-genetic interplay underlying this condition.
Collapse
Affiliation(s)
- Bo-Kyung Shine
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Ja-Eun Choi
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| | - Young-Jin Park
- Department of Family Medicine, Medical Center, Dong-A University, Busan 49201, Republic of Korea
| | - Kyung-Won Hong
- Institute of Advanced Technology, Theragen Health Co., Ltd., Seongnam 13493, Republic of Korea
| |
Collapse
|
10
|
Nakayama A, Kawamura Y, Nakatochi M, Toyoda Y, Nakajima M, Maehara K, Kirihara M, Shimizu S, Matsuo K, Matsuo H. Strong genetic effect on gout revealed by genetic risk score from meta-analysis of two genome-wide association studies. Hum Cell 2024; 38:16. [PMID: 39527284 PMCID: PMC11554751 DOI: 10.1007/s13577-024-01138-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/14/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Mayuko Nakajima
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Kazuki Maehara
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Mana Kirihara
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan.
- Department of Biomedical Information Management, National Defense Medical College Research Institute, National Defense Medical College, Tokorozawa, Japan.
| |
Collapse
|
11
|
Kawamura Y, Nakayama A, Nakatochi M, Aoki Y, Toyoda Y, Nakamura T, Shimizu S, Matsuo K, Shinomiya N, Matsuo H. Identification of novel gout loci from trans-ethnic meta-analysis of serum urate level. Hum Cell 2024; 38:15. [PMID: 39522110 PMCID: PMC11551075 DOI: 10.1007/s13577-024-01128-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuka Aoki
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Takahiro Nakamura
- Laboratory for Mathematics, National Defense Medical College, Tokorozawa, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology & Prevention, Aichi Cancer Center, Nagoya, Japan
- Division of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Japan.
- Department of Biomedical Information Management, National Defense Medical College Research Institute, National Defense Medical College, Tokorozawa, Japan.
| |
Collapse
|
12
|
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
|
13
|
Zeng L, Ma P, Li Z, Liang S, Wu C, Hong C, Li Y, Cui H, Li R, Wang J, He J, Li W, Xiao L, Liu L. Multimodal Machine Learning-Based Marker Enables Early Detection and Prognosis Prediction for Hyperuricemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404047. [PMID: 38976552 PMCID: PMC11425915 DOI: 10.1002/advs.202404047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/19/2024] [Indexed: 07/10/2024]
Abstract
Hyperuricemia (HUA) has emerged as the second most prevalent metabolic disorder characterized by prolonged and asymptomatic period, triggering gout and metabolism-related outcomes. Early detection and prognosis prediction for HUA and gout are crucial for pre-emptive interventions. Integrating genetic and clinical data from 421287 UK Biobank and 8900 Nanfang Hospital participants, a stacked multimodal machine learning model is developed and validated to synthesize its probabilities as an in-silico quantitative marker for hyperuricemia (ISHUA). The model demonstrates satisfactory performance in detecting HUA, exhibiting area under the curves (AUCs) of 0.859, 0.836, and 0.779 within the train, internal, and external test sets, respectively. ISHUA is significantly associated with gout and metabolism-related outcomes, effectively classifying individuals into low- and high-risk groups for gout in the train (AUC, 0.815) and internal test (AUC, 0.814) sets. The high-risk group shows increased susceptibility to metabolism-related outcomes, and participants with intermediate or favorable lifestyle profiles have hazard ratios of 0.75 and 0.53 for gout compared with those with unfavorable lifestyles. Similar trends are observed for other metabolism-related outcomes. The multimodal machine learning-based ISHUA marker enables personalized risk stratification for gout and metabolism-related outcomes, and it is unveiled that lifestyle changes can ameliorate these outcomes within high-risk group, providing guidance for preventive interventions.
Collapse
Affiliation(s)
- Lin Zeng
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pengcheng Ma
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zeyang Li
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shengxing Liang
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chengkai Wu
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
- School of Health Management, Southern Medical University, Guangzhou, 510515, China
| | - Chang Hong
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hao Cui
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ruining Li
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiaren Wang
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jingzhe He
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenyuan Li
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lushan Xiao
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Liu
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
14
|
Leask MP, Crișan TO, Ji A, Matsuo H, Köttgen A, Merriman TR. The pathogenesis of gout: molecular insights from genetic, epigenomic and transcriptomic studies. Nat Rev Rheumatol 2024; 20:510-523. [PMID: 38992217 DOI: 10.1038/s41584-024-01137-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/13/2024]
Abstract
The pathogenesis of gout involves a series of steps beginning with hyperuricaemia, followed by the deposition of monosodium urate crystal in articular structures and culminating in an innate immune response, mediated by the NLRP3 inflammasome, to the deposited crystals. Large genome-wide association studies (GWAS) of serum urate levels initially identified the genetic variants with the strongest effects, mapping mainly to genes that encode urate transporters in the kidney and gut. Other GWAS highlighted the importance of uncommon genetic variants. More recently, genetic and epigenetic genome-wide studies have revealed new pathways in the inflammatory process of gout, including genetic associations with epigenomic modifiers. Epigenome-wide association studies are also implicating epigenomic remodelling in gout, which perhaps regulates the responsiveness of the innate immune system to monosodium urate crystals. Notably, genes implicated in gout GWAS do not include those encoding components of the NLRP3 inflammasome itself, but instead include genes encoding molecules involved in its regulation. Knowledge of the molecular mechanisms underlying gout has advanced through the translation of genetic associations into specific molecular mechanisms. Notable examples include ABCG2, HNF4A, PDZK1, MAF and IL37. Current genetic studies are dominated by participants of European ancestry; however, studies focusing on other population groups are discovering informative population-specific variants associated with gout.
Collapse
Affiliation(s)
- Megan P Leask
- Department of Physiology, University of Otago, Dunedin, Aotearoa, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tania O Crișan
- Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Aichang Ji
- Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Microbiology and Immunology, University of Otago, Dunedin, Aotearoa, New Zealand.
| |
Collapse
|
15
|
Qi H, Sun M, Terkeltaub R, Merriman TR, Chen H, Li Z, Ji A, Xue X, Sun W, Wang C, Li X, He Y, Cui L, Dalbeth N, Li C. Hyperuricemia Subtypes Classified According to Renal Uric Acid Handling Manifesting Distinct Phenotypic and Genetic Profiles in People With Gout. Arthritis Rheumatol 2024; 76:1130-1140. [PMID: 38412854 DOI: 10.1002/art.42838] [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: 06/27/2023] [Revised: 02/19/2024] [Accepted: 02/26/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVE Hyperuricemia can be stratified into four subtypes according to renal uric acid handling. The aim of this study was to comprehensively describe the biologic characteristics (including genetic background) of clinically defined hyperuricemia subtypes in two large geographically independent gout cohorts. METHODS Hyperuricemia subtype was defined as renal uric acid overload (ROL), renal uric acid underexcretion (RUE), combined, or renal normal. Twenty single nucleotide polymorphisms (SNPs) previously identified as gout risk loci or associated with serum urate (SU) concentration in the East Asian population were genotyped. Weighted polygenic risk scores were calculated to assess the cumulative effect of genetic risks on the subtypes. RESULTS Of the 4,873 participants, 8.8% had an ROL subtype, 60.9% RUE subtype, 23.1% combined subtype, and 7.2% normal subtype. The ROL subtype was independently associated with older age at onset, lower SU, tophi, and diabetes mellitus; RUE was associated with lower body mass index (BMI) and non-diabetes mellitus; the combined subtype was associated with younger age at onset, higher BMI, SU, estimated glomerular filtration rate (eGFR), and smoking; and the normal subtype was independently associated with older age at onset, lower SU, and eGFR. Thirteen SNPs were associated with gout with 6 shared loci and subtype-dependent risk loci patterns. High polygenic risk scores were associated with ROL subtype (odds ratio [OR] = 9.63, 95% confidence interval [95% CI] 4.53-15.12), RUE subtype (OR = 2.18, 95% CI 1.57-3.03), and combined subtype (OR = 6.32, 95% CI 4.22-9.48) compared with low polygenic risk scores. CONCLUSION Hyperuricemia subtypes classified according to renal uric acid handling have subtype-specific clinical and genetic features, suggesting subtype-unique pathophysiologic mechanisms.
Collapse
Affiliation(s)
- Han Qi
- The Affiliated Hospital of Qingdao University, Qingdao University, and Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
| | - Mingshu Sun
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Tony R Merriman
- Qingdao University, Qingdao, China, and University of Alabama Birmingham
| | | | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aichang Ji
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaomei Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, and Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
| | - Wenyan Sun
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Can Wang
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinde Li
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuwei He
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingling Cui
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Changgui Li
- The Affiliated Hospital of Qingdao University, Qingdao University, and Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
| |
Collapse
|
16
|
Lee M, Nam S. The causal relationship of serum uric acid on colorectal cancer: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e38722. [PMID: 38941363 PMCID: PMC11466122 DOI: 10.1097/md.0000000000038722] [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: 05/01/2024] [Accepted: 06/06/2024] [Indexed: 06/30/2024] Open
Abstract
Colorectal cancer (CRC) is a significant public health issue owing to its widespread occurrence and substantial morbidity and mortality rates. Recent studies have highlighted serum uric acid (SUA) level as a probable risk factor for CRC; however, the inconsistency in these findings has created doubt. We performed a Mendelian randomization (MR) study utilizing extensive cohort data from the UK BioBank and the NHGRI-EBI Genome-Wide Association Study (GWAS) Catalog to investigate the causal connection between SUA levels and CRC incidence. Our MR study addresses the constraints of earlier studies, including limited sample sizes and inconsistent results. Considering SUA levels as the exposure and CRC as the outcome, the inverse variance-weighted (IVW) approach in MR showed that the odds ratios (ORs) for CRC for each unit increase in SUA were 0.232 (95% confidence interval [CI] of OR 0.094-0.570; P = .001) and 0.551 (95% CI of OR 0.325-0.934; P = .027). Pleiotropic tests and sensitivity analysis confirmed minimal horizontal pleiotropy and the robustness of causality. Our research deepens the understanding of the association between SUA levels and CRC, offering insights into prevention strategies and patient outcomes prediction.
Collapse
Affiliation(s)
- Miseon Lee
- Department of Genome Medicine and Science, AI Convergence Center for Medical Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Seungyoon Nam
- Department of Genome Medicine and Science, AI Convergence Center for Medical Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon, Republic of Korea
| |
Collapse
|
17
|
Bian J, Chen H, Sun J, Han S, Qi M, Pan Q. Retinol dehydrogenase 12 (RDH12) knock out may cause hyperuricemia phenotype in mice. Biochem Biophys Res Commun 2024; 709:149809. [PMID: 38552555 DOI: 10.1016/j.bbrc.2024.149809] [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: 02/27/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Hyperuricemia is a chronic metabolic disease caused by purine metabolism disorder. And several gene loci and transporter proteins that associated with uric acid transport functions have been identified. Retinol Dehydrogenase 12 (RDH12), recognized for its role in safeguarding photoreceptors, and our study investigated the potential impact of Rdh12 mutations on other organs and diseases, particularly hyperuricemia. We assessed Rdh12 mRNA expression levels in various tissues and conducted serum biochemical analyses in Rdh12-/- mice. Compared with the wild type, significant alterations in serum uric acid levels and kidney-related biochemical indicators have been revealed. Then further analysis, including quantitative RT-PCR of gene expression in the liver and kidney, highlighted variations in the expression levels of specific genes linked to hyperuricemia. And renal histology assessment exposed mild pathological lesions in the kidneys of Rdh12-/- mice. In summary, our study suggests that Rdh12 mutations impact not only retinal function but also contribute to hyperuricemia and renal disease phenotypes in mice. Our finding implies that individuals with Rdh12 mutations may be prone to hyperuricemia and gout, emphasizing the significance of preventive measures and regular examinations in daily life.
Collapse
Affiliation(s)
- Jiaxin Bian
- Department of Ophthalmology, Zhejiang University Medical School First Affiliated Hospital, Hangzhou, 310000, China; Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, 310000, China; HVP-China, Hangzhou, 310000, China
| | - Hongyu Chen
- Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, 310000, China
| | - Junhui Sun
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shuai Han
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital(Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310000, China
| | - Ming Qi
- Department of Ophthalmology, Zhejiang University Medical School First Affiliated Hospital, Hangzhou, 310000, China; Department of Cell Biology and Medical Genetics, School of Medicine Zhejiang University, Hangzhou, 310000, China; Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Department of Laboratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310000, China; DIAN Diagnostics, Hangzhou, 310000, China; Department of Pathology and Laboratory of Medicine, University of Rochester Medical Centre, Rochester, NY, 14609, USA; HVP-China, Hangzhou, 310000, China.
| | - Qing Pan
- Department of Ophthalmology, Zhejiang University Medical School First Affiliated Hospital, Hangzhou, 310000, China.
| |
Collapse
|
18
|
Koebbe LL, Hess T, Giel AS, Bigge J, Gehlen J, Schueller V, Geppert M, Dumoulin FL, Heller J, Schepke M, Plaßmann D, Vieth M, Venerito M, Schumacher J, Maj C. The genetic regulation of the gastric transcriptome is associated with metabolic and obesity-related traits and diseases. Physiol Genomics 2024; 56:384-396. [PMID: 38406838 PMCID: PMC11368570 DOI: 10.1152/physiolgenomics.00120.2023] [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: 10/13/2023] [Revised: 01/26/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024] Open
Abstract
Tissue-specific gene expression and gene regulation lead to a better understanding of tissue-specific physiology and pathophysiology. We analyzed the transcriptome and genetic regulatory profiles of two distinct gastric sites, corpus and antrum, to identify tissue-specific gene expression and its regulation. Gastric corpus and antrum mucosa biopsies were collected during routine gastroscopies from up to 431 healthy individuals. We obtained genotype and transcriptome data and performed transcriptome profiling and expression quantitative trait locus (eQTL) studies. We further used data from genome-wide association studies (GWAS) of various diseases and traits to partition their heritability and to perform transcriptome-wide association studies (TWAS). The transcriptome data from corpus and antral mucosa highlights the heterogeneity of gene expression in the stomach. We identified enriched pathways revealing distinct and common physiological processes in gastric corpus and antrum. Furthermore, we found an enrichment of the single nucleotide polymorphism (SNP)-based heritability of metabolic, obesity-related, and cardiovascular traits and diseases by considering corpus- and antrum-specifically expressed genes. Particularly, we could prioritize gastric-specific candidate genes for multiple metabolic traits, like NQO1 which is involved in glucose metabolism, MUC1 which contributes to purine and protein metabolism or RAB27B being a regulator of weight and body composition. Our findings show that gastric corpus and antrum vary in their transcriptome and genetic regulatory profiles indicating physiological differences which are mostly related to digestion and epithelial protection. Moreover, our findings demonstrate that the genetic regulation of the gastric transcriptome is linked to biological mechanisms associated with metabolic, obesity-related, and cardiovascular traits and diseases. NEW & NOTEWORTHY We analyzed the transcriptomes and genetic regulatory profiles of gastric corpus and for the first time also of antrum mucosa in 431 healthy individuals. Through tissue-specific gene expression and eQTL analyses, we uncovered unique and common physiological processes across both primary gastric sites. Notably, our findings reveal that stomach-specific eQTLs are enriched in loci associated with metabolic traits and diseases, highlighting the pivotal role of gene expression regulation in gastric physiology and potential pathophysiology.
Collapse
Affiliation(s)
- Laura L Koebbe
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Timo Hess
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Ann-Sophie Giel
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Jessica Bigge
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - Jan Gehlen
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | | | | | | | - Joerg Heller
- Marienhaus Hospital Ahrweiler, Ahrweiler, Germany
| | - Michael Schepke
- Department of Gastroenterology, Helios Hospital Siegburg, Siegburg, Germany
| | | | - Michael Vieth
- Institute for Pathology, Klinikum Bayreuth, University of Erlangen-Nuremberg, Bayreuth, Germany
| | - Marino Venerito
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital, Magdeburg, Germany
| | | | - Carlo Maj
- Center for Human Genetics, University of Marburg, Marburg, Germany
| |
Collapse
|
19
|
Biswas S, Hilser JR, Woodward NC, Wang Z, Gukasyan J, Nemet I, Schwartzman WS, Huang P, Han Y, Fouladian Z, Charugundla S, Spencer NJ, Pan C, Tang WW, Lusis AJ, Hazen SL, Hartiala JA, Allayee H. Effect of Genetic and Dietary Perturbation of Glycine Metabolism on Atherosclerosis in Humans and Mice. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.08.23299748. [PMID: 38168321 PMCID: PMC10760269 DOI: 10.1101/2023.12.08.23299748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Objective Epidemiological and genetic studies have reported inverse associations between circulating glycine levels and risk of coronary artery disease (CAD). However, these findings have not been consistently observed in all studies. We sought to evaluate the causal relationship between circulating glycine levels and atherosclerosis using large-scale genetic analyses in humans and dietary supplementation experiments in mice. Methods Serum glycine levels were evaluated for association with prevalent and incident CAD in the UK Biobank. A multi-ancestry genome-wide association study (GWAS) meta-analysis was carried out to identify genetic determinants for circulating glycine levels, which were then used to evaluate the causal relationship between glycine and risk of CAD by Mendelian randomization (MR). A glycine feeding study was carried out with atherosclerosis-prone apolipoprotein E deficient (ApoE-/-) mice to determine the effects of increased circulating glycine levels on amino acid metabolism, metabolic traits, and aortic lesion formation. Results Among 105,718 subjects from the UK Biobank, elevated serum glycine levels were associated with significantly reduced risk of prevalent CAD (Quintile 5 vs. Quintile 1 OR=0.76, 95% CI 0.67-0.87; P<0.0001) and incident CAD (Quintile 5 vs. Quintile 1 HR=0.70, 95% CI 0.65-0.77; P<0.0001) in models adjusted for age, sex, ethnicity, anti-hypertensive and lipid-lowering medications, blood pressure, kidney function, and diabetes. A meta-analysis of 13 GWAS datasets (total n=230,947) identified 61 loci for circulating glycine levels, of which 26 were novel. MR analyses provided modest evidence that genetically elevated glycine levels were causally associated with reduced systolic blood pressure and risk of type 2 diabetes, but did provide evidence for an association with risk of CAD. Furthermore, glycine-supplementation in ApoE-/- mice did not alter cardiometabolic traits, inflammatory biomarkers, or development of atherosclerotic lesions. Conclusions Circulating glycine levels were inversely associated with risk of prevalent and incident CAD in a large population-based cohort. While substantially expanding the genetic architecture of circulating glycine levels, MR analyses and in vivo feeding studies in humans and mice, respectively, did not provide evidence that the clinical association of this amino acid with CAD represents a causal relationship, despite being associated with two correlated risk factors.
Collapse
Affiliation(s)
- Subarna Biswas
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - James R. Hilser
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Nicholas C. Woodward
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Zeneng Wang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
- Department of Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Janet Gukasyan
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Ina Nemet
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
- Department of Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195
| | - William S. Schwartzman
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Pin Huang
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Yi Han
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Zachary Fouladian
- Department of Medicine, Immunology, & Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095
| | - Sarada Charugundla
- Department of Medicine, Immunology, & Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095
| | - Neal J. Spencer
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Calvin Pan
- Department of Human Genetics, Immunology, & Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095
| | - W.H. Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
- Department of Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Aldons J. Lusis
- Department of Medicine, Immunology, & Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095
- Department of Human Genetics, Immunology, & Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine of UCLA, Los Angeles, CA 90095
| | - Stanley L. Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
- Department of Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH 44195
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jaana A. Hartiala
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Hooman Allayee
- Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
20
|
Nakayama A, Kurajoh M, Toyoda Y, Takada T, Ichida K, Matsuo H. Dysuricemia. Biomedicines 2023; 11:3169. [PMID: 38137389 PMCID: PMC10740884 DOI: 10.3390/biomedicines11123169] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Gout results from elevated serum urate (SU) levels, or hyperuricemia, and is a globally widespread and increasingly burdensome disease. Recent studies have illuminated the pathophysiology of gout/hyperuricemia and its epidemiology, diagnosis, treatment, and complications. The genetic involvement of urate transporters and enzymes is also proven. URAT1, a molecular therapeutic target for gout/hyperuricemia, was initially derived from research into hereditary renal hypouricemia (RHUC). RHUC is often accompanied by complications such as exercise-induced acute kidney injury, which indicates the key physiological role of uric acid. Several studies have also revealed its physiological role as both an anti-oxidant and a pro-oxidant, acting as both a scavenger and a generator of reactive oxygen species (ROSs). These discoveries have prompted research interest in SU and xanthine oxidoreductase (XOR), an enzyme that produces both urate and ROSs, as status or progression biomarkers of chronic kidney disease and cardiovascular disease. The notion of "the lower, the better" is therefore incorrect; a better understanding of uric acid handling and metabolism/transport comes from an awareness that excessively high and low levels both cause problems. We summarize here the current body of evidence, demonstrate that uric acid is much more than a metabolic waste product, and finally propose the novel disease concept of "dysuricemia" on the path toward "normouricemia", or optimal SU level, to take advantage of the dual roles of uric acid. Our proposal should help to interpret the spectrum from hypouricemia to hyperuricemia/gout as a single disease category.
Collapse
Affiliation(s)
- Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa 359-8513, Japan
| | - Masafumi Kurajoh
- Department of Metabolism, Endocrinology and Molecular Medicine, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa 359-8513, Japan
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Kimiyoshi Ichida
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Science, Hachioji 192-0392, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa 359-8513, Japan
| |
Collapse
|
21
|
Weng H, Li H, Zhang Z, Zhang Y, Xi L, Zhang D, Deng C, Wang D, Chen R, Chen G, Tang S, Zuo X, Yang P, Zhai Z, Wang C. Association between uric acid and risk of venous thromboembolism in East Asian populations: a cohort and Mendelian randomization study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2023; 39:100848. [PMID: 37565068 PMCID: PMC10410163 DOI: 10.1016/j.lanwpc.2023.100848] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/13/2023] [Accepted: 06/29/2023] [Indexed: 08/12/2023]
Abstract
Background Serum uric acid (SUA) levels have been associated with an increased risk and recurrence of venous thromboembolism (VTE) in European populations, but the potential causal relationship remains unclear. Large-scale studies on the association between SUA and VTE in East Asian populations are lacking, despite the high prevalence of hyperuricemia in this region. To address this, we conducted a cohort analysis and a two-sample Mendelian randomization (MR) study in East Asian populations. Methods We collected data on VTE patients from the China Pulmonary Thromboembolism Registry Study (CURES) and compared them to controls obtained from the China Health and Retirement Longitudinal Survey (CHARLS). Propensity score matching (PSM) and cubic-spline models were applied to assess the effect of SUA on VTE risk while adjusting for multiple covariates. We also performed two-sample MR analyses to infer potential causality based on summary statistics from Genome-wide Association Studies (GWAS) of SUA and VTE in the East Asian population. Findings We found that the SUA levels were higher in VTE patients (317.95 mmol/L) compared to the general population (295.75 mmol/L), and SUA ≥ 325 mmol/L was associated with an increased risk of VTE recurrence (P-value = 0.0001). The univariable MR suggested a causal relationship between elevated SUA and higher VTE risk (Pinverse variance weighted < 0.05), and multivariable MR showed that elevated SUA levels continued to promote the development of VTE after adjusting for multiple covariates (Pmultivariable residual < 0.05). Sensitivity analyses produced similar results for these estimations. Interpretation Our study provides evidence supporting a robust positive association between SUA and VTE in the East Asian population, and MR analyses suggest that this association is likely to be causal. Our findings underscore the importance of monitoring SUA levels in VTE prevention and call for urgent action to address the growing burden of hyperuricemia in the Asia-Pacific region. Funding This research was funded by Beijing Nova Program (No. Z211100002121057), National Natural Science Foundation of China (No. 82100065 and No. 62350004), CAMS Innovation Fund for Medical Sciences (No. 2021-I2M-1-061 and No. 2021-1-I2M-001), Elite Medical Professionals project of China-Japan Friendship Hospital (No. ZRJY2021-QM12), National Key Research and Development Project (No. 2021YFF1201200 and No. 2022YFC3341004).
Collapse
Affiliation(s)
- Haoyi Weng
- Shenzhen WeGene Clinical Laboratory, China; WeGene, Shenzhen Zaozhidao Technology Co. Ltd, China; Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, China
| | - Haobo Li
- National Center for Respiratory Medicine, China; State Key Laboratory of Respiratory Health and Multimorbidity, China; National Clinical Research Center for Respiratory Diseases, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Zhu Zhang
- National Center for Respiratory Medicine, China; State Key Laboratory of Respiratory Health and Multimorbidity, China; National Clinical Research Center for Respiratory Diseases, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, China
| | - Yu Zhang
- National Center for Respiratory Medicine, China; State Key Laboratory of Respiratory Health and Multimorbidity, China; National Clinical Research Center for Respiratory Diseases, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, China
- China-Japan Friendship Hospital, Capital Medical University, China
| | - Linfeng Xi
- National Center for Respiratory Medicine, China; State Key Laboratory of Respiratory Health and Multimorbidity, China; National Clinical Research Center for Respiratory Diseases, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, China
- China-Japan Friendship Hospital, Capital Medical University, China
| | - Di Zhang
- Shenzhen WeGene Clinical Laboratory, China; WeGene, Shenzhen Zaozhidao Technology Co. Ltd, China; Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, China
| | - Chao Deng
- SJTU-Yale Joint Center for Biostatistics and Data Science, Shanghai Jiao Tong University, China
| | - Dingyi Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, China; National Center for Respiratory Medicine, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Respiratory Diseases, China
| | - Ruoyan Chen
- Shenzhen WeGene Clinical Laboratory, China; WeGene, Shenzhen Zaozhidao Technology Co. Ltd, China; Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, China
| | - Gang Chen
- Shenzhen WeGene Clinical Laboratory, China; WeGene, Shenzhen Zaozhidao Technology Co. Ltd, China; Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, China
| | - Senwei Tang
- Shenzhen WeGene Clinical Laboratory, China; WeGene, Shenzhen Zaozhidao Technology Co. Ltd, China; Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, China
| | - Xianbo Zuo
- Department of Dermatology, China-Japan Friendship Hospital, China; Department of Pharmacy, China-Japan Friendship Hospital, China
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China; National Center for Respiratory Medicine, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Zhenguo Zhai
- National Center for Respiratory Medicine, China; State Key Laboratory of Respiratory Health and Multimorbidity, China; National Clinical Research Center for Respiratory Diseases, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, China
| | - Chen Wang
- National Center for Respiratory Medicine, China; State Key Laboratory of Respiratory Health and Multimorbidity, China; National Clinical Research Center for Respiratory Diseases, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, China; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, China
| |
Collapse
|
22
|
Deng Y, Huang J, Wong MCS. Association between serum uric acid and prostate cancer risk in East Asian populations: a Mendelian randomization study. Eur J Nutr 2023; 62:1323-1329. [PMID: 36542132 DOI: 10.1007/s00394-022-03076-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Previous observational studies showed that serum uric acid (SUA) was associated with prostate cancer, but the causal relationship is unclear. This study aimed to explore the potential causal association between SUA and prostate cancer risk using Mendelian randomization (MR) analyses in the East Asian populations. METHODS Publicly available summary-level genome-wide association studies (GWAS) data on SUA were obtained from a genome-wide meta-analysis of three Japanese cohorts (121,745 subjects). The GWAS data on prostate cancer were derived from Biobank Japan (109,347 subjects with 5,408 cases and 103,939 controls). A total of 34 SUA-related single-nucleotide polymorphisms (SNPs) (P value < 5 × 10-8) were identified as instrumental variables. The inverse variance weighted method was used as the primary method to compute the odds ratios (ORs) and 95% confidence intervals (95% CIs) for per standard deviation increase in SUA. MR Egger, weighted median, and weighted mode were also applied to test the robustness of the results. RESULTS Genetically predicted SUA was positively associated with prostate cancer risk using inverse variance weighted (OR = 1.12; 95% CI 1.00-1.26; P = 0.043). The positive association was robust when MR Egger (OR = 1.16; 95% CI 1.01-1.34; P = 0.048), weighted median (OR = 1.18; 95% CI 1.03-1.36; P = 0.018), and weighted mode (OR = 1.14; 95% CI 1.01-1.29; P = 0.041) were used. CONCLUSION There were potential causal associations between higher genetically predicted SUA levels and increased prostate cancer risk. Further, MR studies with more valid SNPs and more cancer cases are needed. Validation of the findings is also recommended.
Collapse
Affiliation(s)
- Yunyang Deng
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Martin Chi Sang Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR, 999077, China.
- School of Public Health, the Chinese Academy of Medical Sciences and the Peking Union Medical College, Beijing, 100000, China.
- School of Public Health, Peking University, Beijing, 100000, China.
| |
Collapse
|
23
|
Fernández-Torres J, Zamudio-Cuevas Y, Martínez-Nava GA, Martínez-Flores K, Ruíz-Dávila X, Sánchez-Sánchez R. Relationship between rs4349859 and rs116488202 polymorphisms close to MHC-I region and serum urate levels in patients with gout. Mol Biol Rep 2023; 50:4367-4374. [PMID: 36943604 DOI: 10.1007/s11033-023-08359-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/23/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Gout is the most common inflammatory rheumatic disease and elevated levels of serum urate (SU) are the main cause for its development. Major histocompatibility complex class 1 (MHC-1) plays an important role in the development of multiple inflammatory diseases; however, there is little evidence of its involvement in gout. The present study focused on evaluating the association of the rs4349859 and rs116488202 single nucleotide polymorphisms (SNPs) close to the MHC-1 region in patients with gout. METHODS AND RESULTS One hundred and seventy-six individuals of Mexican origin were included, of which 81 were patients with primary gout and 95 were healthy controls. The rs4349859 and rs116488202 SNPs were genotyped using TaqMan probes by allelic discrimination by real-time PCR. Serum concentrations of biochemical parameters were measured with enzymatic methods. Descriptive statistics were applied and P-values < 0.05 were considered significant. It was observed that the rs4349859 and rs116488202 SNPs showed significant association with the risk of gout (OR = 146, 95%CI = 44.8-480.2, P < 0.01; OR = 2885, 95%CI = 265-31398, P < 0.01, respectively). Our results also showed significantly higher serum SU levels in gout patients with respect to controls (P < 0.01) in the carriers of the GA genotype compared with the GG genotype of the rs4349859 variant, and in the carriers of the CT genotype compared with the CC genotype of the rs116488202 variant. CONCLUSION The study revealed that rs4349859 and rs116488202 SNPs close to MHC-I region confers strong susceptibility to gout in Mexican population, and the heterozygous genotypes of both were associated with higher levels of SU.
Collapse
Affiliation(s)
- Javier Fernández-Torres
- Synovial Fluid Laboratory, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico.
- Biology Department, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.
| | - Yessica Zamudio-Cuevas
- Synovial Fluid Laboratory, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | | | - Karina Martínez-Flores
- Synovial Fluid Laboratory, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | | | - Roberto Sánchez-Sánchez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| |
Collapse
|
24
|
Xu L, Cheng J, Lu J, Lin G, Yu Q, Li Y, Chen J, Xie J, Su Z, Zhou Q. Integrating network pharmacology and experimental validation to clarify the anti-hyperuricemia mechanism of cortex phellodendri in mice. Front Pharmacol 2022; 13:964593. [PMID: 36438835 PMCID: PMC9692208 DOI: 10.3389/fphar.2022.964593] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/24/2022] [Indexed: 08/12/2023] Open
Abstract
Hyperuricemia (HUA), a common metabolic disease, is treated as the second-largest metabolic disease after diabetes in China. Cortex Phellodendri (CP) is one of the most frequently used herbal medicines for treating gout or HUA. However, the mechanism underlying the anti-HUA effect of CP is still unrevealed. Hence, this study aimed to explore the pharmacological mechanism of CP against HUA using network pharmacology coupled with in vivo experimental validation. Active compounds and potential targets of CP, as well as the potential targets related to HUA, were retrieved from multiple open-source databases. The drug-disease overlapping targets were obtained by Venn diagram analysis and used to construct the herb-component-target (HCT), protein-protein-interaction (PPI), and component-target-pathway (CTP) networks. The functional enrichment analysis was also performed for further study. Furthermore, a HUA mouse model was induced by a combination of intraperitoneal injection of potassium oxonate (PO, 300 mg/kg) and intragastric administration of hypoxanthine (HX, 300 mg/kg) daily for 10 days. Different dosages of CP (200, 400, and 800 mg/kg) were orally given to mice 1 h after modeling. The results showed that 12 bioactive compounds and 122 drug-disease overlapping targets were obtained by matching 415 CP-related targets and 679 HUA-related targets, and berberine was one of the most important compounds with the highest degree value. The core targets of CP for treating HUA were TP53, MAPK8, MAPK3, IL-6, c-Jun, AKT1, xanthine oxidase (XOD), and ATP-binding cassette subfamily G member 2 (ABCG2). The Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment results showed that the anti-HUA effect of CP mainly involved the pathways of inflammation and apoptosis, such as PI3K/Akt, TNF, MAPK, TLR, AMPK, NF-κB, and NLRP3 signaling pathways. In vivo animal experiment further confirmed the hypouricemic effect of CP in a HUA mouse model, as evidenced by significantly restored kidney histological deteriorations, and considerably decreased levels of serum uric acid (sUA), creatinine (Cre), blood urea nitrogen (BUN), and hepatic UA. Furthermore, the hypouricemic action of CP in vivo might be attributed to its suppression of XOD activity in the liver, rather than ABCG2 in the kidney. Real-time qPCR (RT-qPCR) and Western blot analysis also confirmed the key roles of the hub genes in CP against HUA. In conclusion, CP exhibited therapeutic effect against HUA via multi-compounds, multi-targets, and multi-pathways. It possessed anti-HUA and nephroprotective effects via suppressing XOD activity, and reversed the progression of renal injury by exerting anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Lieqiang Xu
- College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, China
| | - Juanjuan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jieyi Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiuxia Yu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| |
Collapse
|
25
|
Lai B, Yu HP, Chang YJ, Wang LC, Chen CK, Zhang W, Doherty M, Chang SH, Hsu JT, Yu KH, Kuo CF. Assessing the causal relationships between gout and hypertension: a bidirectional Mendelian randomisation study with coarsened exposures. Arthritis Res Ther 2022; 24:243. [PMID: 36309757 PMCID: PMC9617405 DOI: 10.1186/s13075-022-02933-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 10/20/2022] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES Observational studies have demonstrated associations between gout and hypertension, but whether they are causal remains unclear. Our work aims to assess the causal relationship between gout and hypertension. METHODS We obtained genetic information from the Taiwan Biobank, including 88,347 participants and 686,439 single-nucleotide polymorphisms (SNPs). A novel model of Mendelian randomisation (MR) with coarsened exposures was used to examine the causality between the liability of gout on hypertension and vice versa, using 4 SNPs associated with gout and 10 SNPs associated with hypertension after removal of SNPs associated with measured confounders. The binary exposure (gout/hypertension) can be considered a coarsened approximation of a latent continuous trait. The inverse-variance weighted (IVW) and polygenic risk score (PRS) methods were used to estimate effect size. The MR analysis with coarsened exposures was performed with and without adjustments for covariates. RESULTS Of the 88,347 participants, 3253 (3.68%) had gout and 11,948 (13.52%) had hypertension (men, 31.9%; mean age 51.1 [SD, 11.1] years). After adjusting to measured confounders, MR analysis with coarsened exposures showed a significant positive causal effect of the liability of gout on hypertension in both the IVW method (relative risk [RR], 1.10; 95% confidence interval [CI], 1.03-1.19; p = 0.0077) and the PRS method (RR, 1.10; 95% CI, 1.02-1.19; p = 0.0092). The result of causality was the same before and after involving measured confounders. However, there was no causal effect of the liability of hypertension on gout. CONCLUSIONS In this study, we showed that the liability of gout has a causal effect on hypertension, but the liability of hypertension does not have a causal effect on gout. Adequate management of gout may reduce the risk of developing hypertension.
Collapse
Affiliation(s)
- Benjamin Lai
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Jing Chang
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Liang-Chin Wang
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Che-Kai Chen
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Weiya Zhang
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, UK
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, UK
| | - Michael Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, UK
- Pain Centre Versus Arthritis, University of Nottingham, Nottingham, UK
| | - Shang-Hung Chang
- Division of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan City, Taiwan
- Center for Big Data Analytics and Statistics, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- Graduate Institute of Nursing, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Jun-Te Hsu
- Department of General Surgery, Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Chang, Taiwan
| | - Kuang-Hui Yu
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chang-Fu Kuo
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
26
|
Wang K, Cadzow M, Bixley M, Leask MP, Merriman ME, Yang Q, Li Z, Takei R, Phipps-Green A, Major TJ, Topless R, Dalbeth N, King F, Murphy R, Stamp LK, de Zoysa J, Wang Z, Shi Y, Merriman TR. A Polynesian-specific copy number variant encompassing the MICA gene associates with gout. Hum Mol Genet 2022; 31:3757-3768. [PMID: 35451026 PMCID: PMC9616569 DOI: 10.1093/hmg/ddac094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/01/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Gout is of particularly high prevalence in the Māori and Pacific (Polynesian) populations of Aotearoa New Zealand (NZ). Here, we investigated the contribution of common population-specific copy number variation (CNV) to gout in the Aotearoa NZ Polynesian population. Microarray-generated genome-wide genotype data from Aotearoa NZ Polynesian individuals with (n = 1196) and without (n = 1249) gout were analyzed. Comparator population groups were 552 individuals of European ancestry and 1962 of Han Chinese ancestry. Levels of circulating major histocompatibility complex (MHC) class I polypeptide-related sequence A (MICA) were measured by enzyme-linked immunosorbent assay. Fifty-four CNV regions (CNVRs) appearing in at least 10 individuals were detected, of which seven common (>2%) CNVRs were specific to or amplified in Polynesian people. A burden test of these seven revealed associations of insertion/deletion with gout (odds ratio (OR) 95% confidence interval [CI] = 1.80 [1.01; 3.22], P = 0.046). Individually testing of the seven CNVRs for association with gout revealed nominal association of CNVR1 with gout in Western Polynesian (Chr6: 31.36-31.45 Mb, OR = 1.72 [1.03; 2.92], P = 0.04), CNVR6 in the meta-analyzed Polynesian sample sets (Chr1: 196.75-196.92 Mb, OR = 1.86 [1.16; 3.00], P = 0.01) and CNVR9 in Western Polynesian (Chr1: 189.35-189.54 Mb, OR = 2.75 [1.15; 7.13], P = 0.03). Analysis of European gout genetic association data demonstrated a signal of association at the CNVR1 locus that was an expression quantitative trait locus for MICA. The most common CNVR (CNVR1) includes deletion of the MICA gene, encoding an immunomodulatory protein. Expression of MICA was reduced in the serum of individuals with the deletion. In summary, we provide evidence for the association of CNVR1 containing MICA with gout in Polynesian people, implicating class I MHC-mediated antigen presentation in gout.
Collapse
Affiliation(s)
- Ke Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Murray Cadzow
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Matt Bixley
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Megan P Leask
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Marilyn E Merriman
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Qiangzhen Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
| | - Zhiqiang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
- Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao 266003, China
| | - Riku Takei
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | - Tanya J Major
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Ruth Topless
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland 1023, New Zealand
| | - Frances King
- Ngati Porou Hauora Charitable Trust, Te Puia Springs, New Zealand
| | - Rinki Murphy
- Department of Medicine, University of Auckland, Auckland 1023, New Zealand
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch 8013, New Zealand
| | - Janak de Zoysa
- Department of Medicine, University of Auckland, Auckland 1023, New Zealand
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, People’s Republic of China
- Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao 266003, China
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
27
|
Zhang X, Mass BB, Talevi V, Hou R, North KE, Voruganti VS. Novel Insights into the Effects of Genetic Variants on Serum Urate Response to an Acute Fructose Challenge: A Pilot Study. Nutrients 2022; 14:4030. [PMID: 36235682 PMCID: PMC9570712 DOI: 10.3390/nu14194030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 11/25/2022] Open
Abstract
Studies have shown that genetic variations can influence metabolic response to nutrient intake, and that diets rich in fructose contribute to hyperuricemia. In this pilot study, our aim was to determine the variability of serum urate in response to an acute fructose challenge and to investigate if genetic variants would affect this response in young to middle-aged adults who self-reported as Black or White. Fifty-seven participants consumed a fructose-rich beverage after an overnight fast. Blood was drawn at five time points (baseline, 30, 60, 120, and 180 min after consumption). Thirty urate-related single nucleotide polymorphisms (SNPs) were analyzed for their associations with baseline serum urate and its percent changes, using a two-step modeling approach followed by meta-analysis. At baseline, serum urate (mg/dL, mean ± SD) was higher in Whites (5.60 ± 1.01 vs. 5.37 ± 0.96), men (6.17 ± 1.14 vs. 5.24 ± 0.79), and those with obesity (5.69 ± 1.08 vs. 5.42 ± 1.06 vs. 5.34 ± 0.80). Three SNPs were significantly associated with baseline serum urate or its percent changes, and six SNPs were nominally associated with percent changes in serum urate. In summary, our results showed that genetic variants could play a role in short-term urate metabolism.
Collapse
Affiliation(s)
- Xinruo Zhang
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Baba B Mass
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Valentina Talevi
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Ruixue Hou
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Venkata Saroja Voruganti
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
28
|
Zhao J, Guo S, Schrodi SJ, He D. Trends in the Contribution of Genetic Susceptibility Loci to Hyperuricemia and Gout and Associated Novel Mechanisms. Front Cell Dev Biol 2022; 10:937855. [PMID: 35813212 PMCID: PMC9259951 DOI: 10.3389/fcell.2022.937855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/31/2022] [Indexed: 11/14/2022] Open
Abstract
Hyperuricemia and gout are complex diseases mediated by genetic, epigenetic, and environmental exposure interactions. The incidence and medical burden of gout, an inflammatory arthritis caused by hyperuricemia, increase every year, significantly increasing the disease burden. Genetic factors play an essential role in the development of hyperuricemia and gout. Currently, the search on disease-associated genetic variants through large-scale genome-wide scans has primarily improved our understanding of this disease. However, most genome-wide association studies (GWASs) still focus on the basic level, whereas the biological mechanisms underlying the association between genetic variants and the disease are still far from well understood. Therefore, we summarized the latest hyperuricemia- and gout-associated genetic loci identified in the Global Biobank Meta-analysis Initiative (GBMI) and elucidated the comprehensive potential molecular mechanisms underlying the effects of these gene variants in hyperuricemia and gout based on genetic perspectives, in terms of mechanisms affecting uric acid excretion and reabsorption, lipid metabolism, glucose metabolism, and nod-like receptor pyrin domain 3 (NLRP3) inflammasome and inflammatory pathways. Finally, we summarized the potential effect of genetic variants on disease prognosis and drug efficacy. In conclusion, we expect that this summary will increase our understanding of the pathogenesis of hyperuricemia and gout, provide a theoretical basis for the innovative development of new clinical treatment options, and enhance the capabilities of precision medicine for hyperuricemia and gout treatment.
Collapse
Affiliation(s)
- Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of WI-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of WI-Madison, Madison, WI, United States
| | - Steven J. Schrodi
- Computation and Informatics in Biology and Medicine, University of WI-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of WI-Madison, Madison, WI, United States
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
29
|
Park JS, Kim Y, Kang J. Genome-wide meta-analysis revealed several genetic loci associated with serum uric acid levels in Korean population: an analysis of Korea Biobank data. J Hum Genet 2022; 67:231-237. [PMID: 34719683 DOI: 10.1038/s10038-021-00991-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022]
Abstract
The serum uric acid (SUA) level is an important determinant of gout, hypertension, metabolic syndrome, and cardiovascular disease. Although previous genome-wide studies have identified multiple genetic variants associated with SUA, most genetic analyses have focused on individuals with European ancestry; thus, understanding of the genetic architecture of SUA is currently limited for Asian populations. We conducted a genome-wide meta-analysis based on Korea Biobank data consistent with three cohorts; namely, the Korean Genome and Epidemiology Study (KoGES) Ansan and Ansung, KoGES Health Examinee, and KoGES Cardiovascular Disease Association studies. In total, 60,585 participants aged ≥40 years were included in the analysis of the three cohorts. We used logistic regression analyses to perform genome-wide association study (GWAS) adjustments for confounding variables. Subsequently, a meta-analysis was conducted by combining the analyses of the three GWASs. We identified 8,105 variants at 22 genetic loci with a P value < 5 × 10-8. Among these, six novel genetic loci associated with SUA in the Korean population were identified (rs4715517 in HCRTR2, rs145099458 in 3.2 kb 3' of MLXIPL, rs1137642 in B4GALT1, rs659107 in LOC105378410, rs7919329 in LOC107984274, and rs2240751 in MFSD12). Our meta-analysis provides insights into the genetic architecture of SUA in the Korean population. Further studies are warranted to replicate the study results and elucidate the specific role of these variants in SUA homeostasis.
Collapse
Affiliation(s)
- Jin Sung Park
- Department of Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
| | - Yunkyung Kim
- Division of Rheumatology, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
| | - Jihun Kang
- Department of Family Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
30
|
Shirai Y, Nakayama A, Kawamura Y, Toyoda Y, Nakatochi M, Shimizu S, Shinomiya N, Okada Y, Matsuo H. Coffee Consumption Reduces Gout Risk Independently of Serum Uric Acid Levels: Mendelian Randomization Analyses Across Ancestry Populations. ACR Open Rheumatol 2022; 4:534-539. [PMID: 35348303 PMCID: PMC9190218 DOI: 10.1002/acr2.11425] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/16/2022] [Accepted: 01/26/2022] [Indexed: 12/02/2022] Open
Abstract
Objective The effects of coffee consumption on serum uric acid (SUA) levels and gout risk are controversial. There have hitherto been no reports based on Mendelian randomization (MR) analysis of its effects that consider pleiotropy. Here, we evaluated the effects of coffee consumption across ancestry populations, taking pleiotropy into account. Methods We performed the first MR analyses for coffee consumption on SUA levels and gout, considering pleiotropy. We used the following summary statistics of genome‐wide association studies from a Japanese population: habitual coffee consumption (152,634 subjects), gout (3053 cases and 4554 controls), and SUA levels (121,745 subjects). In addition to fixed‐effect inverse variance weighted (IVW) meta‐analysis, we performed a robust evaluation of heterogeneity and removed several instruments for reasons of possible pleiotropy. Previous European datasets were also reevaluated while heterogeneity was considered. Results Habitual coffee consumption was significantly and inversely associated with gout (odds ratio [OR] = 0.29, 95% confidence interval [95% CI]: 0.16‐0.51, P = 1.9 × 10−5) in random‐effect IVW (Phet = 5.5 × 10−19). Excluding pleiotropic instruments, the protective effect on gout was confirmed in fixed‐effect IVW analysis (OR = 0.75, 95% CI: 0.58‐0.97, P = 0.026) without heterogeneity (Phet = 0.39). However, we observed no significance in the previous European datasets when heterogeneity was considered. Associations were not observed between coffee consumption and SUA levels in either ancestry in MR analyses that considered pleiotropy. Multivariable MR analysis showed that increased coffee consumption significantly reduced gout risk, even after adjusting for SUA levels (OR = 0.50, 95% CI: 0.31‐0.81, P = 0.0046). Conclusion With pleiotropy taken into account, our MR analyses revealed that coffee consumption can causally reduce gout risk, and that it may reduce gout risk independently of SUA levels.
Collapse
Affiliation(s)
- Yuya Shirai
- Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | - Yu Toyoda
- National Defense Medical College, Tokorozawa, Japan
| | | | | | | | - Yukinori Okada
- Osaka University Graduate School of Medicine and Osaka University, Suita, Japan, and RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | | |
Collapse
|
31
|
Sun M, Sun W, Zhao X, Li Z, Dalbeth N, Ji A, He Y, Qu H, Zheng G, Ma L, Wang J, Shi Y, Fang X, Chen H, Merriman TR, Li C. A machine learning-assisted model for renal urate underexcretion with genetic and clinical variables among Chinese men with gout. Arthritis Res Ther 2022; 24:67. [PMID: 35264217 PMCID: PMC8905745 DOI: 10.1186/s13075-022-02755-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 02/28/2022] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES The objective of this study was to develop and validate a prediction model for renal urate underexcretion (RUE) in male gout patients. METHODS Men with gout enrolled from multicenter cohorts in China were analyzed as the development and validation data sets. The RUE phenotype was defined as fractional excretion of uric acid (FEUA) <5.5%. Candidate genetic and clinical features were screened by the least absolute shrinkage and selection operator (LASSO) with 10-fold cross-validation. Machine learning algorithms (stochastic gradient descent (SGD), logistic regression, support vector machine) were performed to construct a predictive classifier of RUE. Models were assessed by the area under the receiver operating characteristic curve (AUC) and the precision-recall curve (PRC). RESULTS One thousand two hundred thirty-eight and two thousand twenty-three patients were enrolled as the development and validation cohorts, with 1220 and 754 randomly chosen patients genotyped, respectively. Rs3775948.GG of SLC2A9/GLUT9, rs504915.AA of NRXN2/URAT1, and 7 clinical features (age, hypertension, nephrolithiasis, blood glucose, serum urate, urea nitrogen, and creatinine) were generated by LASSO. Two additional SNP variants (rs2231142.GG of ABCG2 and rs11231463.GG of SLC22A9/OAT7) were selected based on their contributions to gout in the development cohort and their reported effects on renal urate handling. The optimized classifiers yielded AUCs of ~0.914 and PRCs of ~0.980 using these 11 variables. The SGD model was conducted in the validation cohort with an AUC of 0.899 and the PRC of 0.957. CONCLUSIONS A prediction model for RUE composed of four SNPs and readily accessible clinical features was established with acceptable accuracy for men with gout.
Collapse
Affiliation(s)
- Mingshu Sun
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenyan Sun
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuetong Zhao
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Zhiqiang Li
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Aichang Ji
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuwei He
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongzhu Qu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Guangmin Zheng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Lidan Ma
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiayi Wang
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Yongyong Shi
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangdong Fang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Haibing Chen
- Department of Endocrinology and Metabolism, Shanghai 10th People's Hospital, Tongji University, Shanghai, China.
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand. .,Department of Medicine, University of Alabama Birmingham, Birmingham, AL, USA.
| | - Changgui Li
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
32
|
Copula-Based Estimation Methods for a Common Mean Vector for Bivariate Meta-Analyses. Symmetry (Basel) 2022. [DOI: 10.3390/sym14020186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Traditional bivariate meta-analyses adopt the bivariate normal model. As the bivariate normal distribution produces symmetric dependence, it is not flexible enough to describe the true dependence structure of real meta-analyses. As an alternative to the bivariate normal model, recent papers have adopted “copula” models for bivariate meta-analyses. Copulas consist of both symmetric copulas (e.g., the normal copula) and asymmetric copulas (e.g., the Clayton copula). While copula models are promising, there are only a few studies on copula-based bivariate meta-analysis. Therefore, the goal of this article is to fully develop the methodologies and theories of the copula-based bivariate meta-analysis, specifically for estimating the common mean vector. This work is regarded as a generalization of our previous methodological/theoretical studies under the FGM copula to a broad class of copulas. In addition, we develop a new R package, “CommonMean.Copula”, to implement the proposed methods. Simulations are performed to check the proposed methods. Two real dataset are analyzed for illustration, demonstrating the insufficiency of the bivariate normal model.
Collapse
|
33
|
Chang SJ, Toyoda Y, Kawamura Y, Nakamura T, Nakatochi M, Nakayama A, Liao WT, Shimizu S, Takada T, Takeuchi K, Wakai K, Shi Y, Shinomiya N, Chen CJ, Li C, Okada Y, Ichida K, Matsuo H. A meta-analysis of genome-wide association studies using Japanese and Taiwanese has revealed novel loci associated with gout susceptibility. Hum Cell 2022; 35:767-770. [PMID: 35032298 PMCID: PMC8866370 DOI: 10.1007/s13577-021-00665-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/21/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Shun-Jen Chang
- Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, Kaohsiung, Taiwan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takahiro Nakamura
- Laboratory for Mathematics, National Defense Medical College, Saitama, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Wei-Ting Liao
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Kenji Takeuchi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Chung-Jen Chen
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Changgui Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kimiyoshi Ichida
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| |
Collapse
|
34
|
Chen Y, Huang T, Yu Z, Yu Q, Wang Y, Hu J, Shi J, Yang G. The functions and roles of sestrins in regulating human diseases. Cell Mol Biol Lett 2022; 27:2. [PMID: 34979914 PMCID: PMC8721191 DOI: 10.1186/s11658-021-00302-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Sestrins (Sesns), highly conserved stress-inducible metabolic proteins, are known to protect organisms against various noxious stimuli including DNA damage, oxidative stress, starvation, endoplasmic reticulum (ER) stress, and hypoxia. Sesns regulate metabolism mainly through activation of the key energy sensor AMP-dependent protein kinase (AMPK) and inhibition of mammalian target of rapamycin complex 1 (mTORC1). Sesns also play pivotal roles in autophagy activation and apoptosis inhibition in normal cells, while conversely promoting apoptosis in cancer cells. The functions of Sesns in diseases such as metabolic disorders, neurodegenerative diseases, cardiovascular diseases, and cancer have been broadly investigated in the past decades. However, there is a limited number of reviews that have summarized the functions of Sesns in the pathophysiological processes of human diseases, especially musculoskeletal system diseases. One aim of this review is to discuss the biological functions of Sesns in the pathophysiological process and phenotype of diseases. More significantly, we include some new evidence about the musculoskeletal system. Another purpose is to explore whether Sesns could be potential biomarkers or targets in the future diagnostic and therapeutic process.
Collapse
Affiliation(s)
- Yitong Chen
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Tingben Huang
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Zhou Yu
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Qiong Yu
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Ying Wang
- Department of Oral Medicine, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Ji'an Hu
- Department of Oral Pathology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| | - Jiejun Shi
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| | - Guoli Yang
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
35
|
Ikezaki H, Furusyo N, Nakashima R, Umemoto M, Yamamoto K, Matsumoto Y, Ohta A, Yamasaki S, Hiramine S, Takayama K, Ogawa E, Toyoda K, Murata M, Shimono N, Hayashi J. Kyushu and Okinawa Population Study (KOPS): a large prospective cohort study in Japan. BMJ Open 2021; 11:e053763. [PMID: 34933862 PMCID: PMC8693098 DOI: 10.1136/bmjopen-2021-053763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The Kyushu and Okinawa Population Study (KOPS) was established to investigate gene-environmental interactions in non-communicable diseases in Japan. Besides collecting blood samples and anthropometric measurements, we also obtained medical histories, psychological status and lifestyle habits, including physical activities and dietary patterns. PARTICIPANTS KOPS is a community-based prospective cohort study and consists of participants from four southwestern areas in Japan. Baseline surveys were conducted between 2004 and 2007 (wave 1), and 2009 and 2012 (wave 2) at the sites of municipality-based health check-ups. A total of 17 077 participants were included, comprising 10 697 participants of wave 1 and 6380 participants of wave 2; the median age in both groups was 61 years. Among them, 3006 individuals participated in both wave 1 and wave 2 surveys. FINDINGS TO DATE We have focused on either risk or confounding factors for non-communicable diseases. We have assessed the clinical utility of the newly developed biomarkers for impaired glucose tolerance, such as urinary myo-inositol and glycated albumin, and atherosclerosis, such as small dense low-density lipoprotein cholesterol. We have conducted an international collaborative study with Framingham Offspring Study to investigate ethnic differences in impaired glucose tolerance and cardiovascular diseases. We have found that insulin resistance and deficiency might account for the ethnic differences in impaired glucose tolerance and cardiovascular disease risks. As gene-environmental interaction analyses, we found a synergic effect of interleukin 28B single nucleotide polymorphisms (SNPs) and gender on the spontaneous elimination of hepatitis C, and a beneficial interaction of SNPs of high-density lipoprotein cholesterol and gender on the impact of physical activity. In addition, we reported eight novel loci contributing to the development and severity of coronary artery disease from a large genome-wide association study. FUTURE PLANS We plan to investigate further the clinical utility of the newly developed biomarkers and the gene-environmental interactions using prospective data.
Collapse
Affiliation(s)
- Hiroaki Ikezaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Comprehensive General Internal Medicine, Kyushu University Faculty of Medical Sciences, Fukuoka, Japan
| | - Norihiro Furusyo
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Taihaku Avenue Clinic, Fukuoka, Japan
| | - Ryoko Nakashima
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Makiko Umemoto
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Ken Yamamoto
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Yuji Matsumoto
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Environmental Medicine and Infectious Diseases, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Azusa Ohta
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Environmental Medicine and Infectious Diseases, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Sho Yamasaki
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Environmental Medicine and Infectious Diseases, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Satoshi Hiramine
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Koji Takayama
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Eiichi Ogawa
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Environmental Medicine and Infectious Diseases, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Kazuhiro Toyoda
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Masayuki Murata
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Nobuyuki Shimono
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Jun Hayashi
- Department of General Internal Medicine, Kyushu University Hospital, Fukuoka, Japan
- Kyushu General Internal Medicine Center, Haradoi Hospital, Fukuoka, Japan
| |
Collapse
|
36
|
Abstract
Circulation of urate levels is determined by the balance between urate production and excretion, homeostasis regulated by the function of urate transporters in key epithelial tissues and cell types. Our understanding of these physiological processes and identification of the genes encoding the urate transporters has advanced significantly, leading to a greater ability to predict risk for urate-associated diseases and identify new therapeutics that directly target urate transport. Here, we review the identified urate transporters and their organization and function in the renal tubule, the intestinal enterocytes, and other important cell types to provide a fuller understanding of the complicated process of urate homeostasis and its role in human diseases. Furthermore, we review the genetic tools that provide an unbiased catalyst for transporter identification as well as discuss the role of transporters in determining the observed significant gender differences in urate-associated disease risk.
Collapse
Affiliation(s)
| | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
37
|
Ogura M, Toyoda Y, Sakiyama M, Kawamura Y, Nakayama A, Yamanashi Y, Takada T, Shimizu S, Higashino T, Nakajima M, Naito M, Hishida A, Kawai S, Okada R, Sasaki M, Ayaori M, Suzuki H, Takata K, Ikewaki K, Harada-Shiba M, Shinomiya N, Matsuo H. Increase of serum uric acid levels associated with APOE ε2 haplotype: a clinico-genetic investigation and in vivo approach. Hum Cell 2021; 34:1727-1733. [PMID: 34532841 PMCID: PMC8490264 DOI: 10.1007/s13577-021-00609-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/02/2021] [Indexed: 10/30/2022]
Abstract
Elevated serum uric acid (SUA)-hyperuricemia-is caused by overproduction of urate or by its decreased renal and/or intestinal excretion. This disease, which is increasing in prevalence worldwide, is associated with both gout and metabolic diseases. Several studies have reported relationships between apolipoprotein E (APOE) haplotypes and SUA levels in humans; however, their results remain inconsistent. This prompted us to investigate the relationship between APOE polymorphisms and SUA levels. Our subjects were 5,272 Japanese men, premenopausal women, and postmenopausal women. Multiple linear regression analyses revealed the ε2 haplotype of APOE to be independently associated with higher SUA in men (N = 1,726) and postmenopausal women (N = 1,753), but not in premenopausal women (N = 1,793). In contrast, the ε4 haplotype was little related to SUA levels in each group. Moreover, to examine the effect of Apoe deficiency on SUA levels, we conducted animal experiments using Apoe knockout mice, which mimics ε2/ε2 carriers. We found that SUA levels in Apoe knockout mice were significantly higher than those in wild-type mice, which is consistent with the SUA-raising effect of the ε2 haplotype observed in our clinico-genetic analyses. Further analyses suggested that renal rather than intestinal underexcretion of urate could be involved in Apoe deficiency-related SUA increase. In conclusion, we successfully demonstrated that the ε2 haplotype, but not the ε4 haplotype, increases SUA levels. These findings will improve our understanding of genetic factors affecting SUA levels.
Collapse
Affiliation(s)
- Masatsune Ogura
- Department of Metabolism and Endocrinology, Eastern Chiba Medical Center, 3-6-2 Okayamadai, Togane, Chiba, 283-8686, Japan.
- Department of General Medical Science, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8670, Japan.
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, Osaka, 564-8565, Japan.
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Masayuki Sakiyama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
- Department of Dermatology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yoshihide Yamanashi
- Department of Pharmacy, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tappei Takada
- Department of Pharmacy, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Toshihide Higashino
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Mayuko Nakajima
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
- Department of Oral Epidemiology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Sayo Kawai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
- Department of Public Health, Aichi Medical University School of Medicine, 1-1 Yazako-karimata, Nagakute, Aichi, 480-1195, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Makoto Sasaki
- Division of Anti-Aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Makoto Ayaori
- Division of Anti-Aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Koki Takata
- Takata Clinic, 10-15 Wakakusa-cho, Higashi-ku, Hiroshima, Hiroshima, 732-0053, Japan
| | - Katsunori Ikewaki
- Division of Anti-Aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Mariko Harada-Shiba
- Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, Osaka, 564-8565, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| |
Collapse
|
38
|
The genetic basis of urate control and gout: Insights into molecular pathogenesis from follow-up study of genome-wide association study loci. Best Pract Res Clin Rheumatol 2021; 35:101721. [PMID: 34732286 DOI: 10.1016/j.berh.2021.101721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review focuses on the post-genome-wide association study (GWAS) era in gout, i.e., the translation of GWAS genetic association signals into biologically informative knowledge. Analytical and experimental follow-up of individual loci, based on the identification of causal genetic variants, reveals molecular pathogenic pathways. We summarize in detail the largest GWAS in urate to date, then we review follow-up studies and molecular insights from ABCG2, HNF4A, PDZK1, MAF, GCKR, ALDH2, ALDH16A1, SLC22A12, SLC2A9, ABCC4, and SLC22A13, including the role of insulin signaling. One common factor in these pathways is the importance of transcriptional control, including the HNF4α transcription factor. The new molecular knowledge reveals new targets for intervention to manage urate levels and prevent gout.
Collapse
|
39
|
Pleiotropic Effects of Functional MUC1 Variants on Cardiometabolic, Renal, and Hematological Traits in the Taiwanese Population. Int J Mol Sci 2021; 22:ijms221910641. [PMID: 34638981 PMCID: PMC8509060 DOI: 10.3390/ijms221910641] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/09/2021] [Accepted: 09/27/2021] [Indexed: 12/27/2022] Open
Abstract
MUC1 is a transmembrane mucin involved in carcinogenesis and cell signaling. Functional MUC1 variants are associated with multiple metabolic and biochemical traits. This study investigated the association of functional MUC1 variants with MUC1 DNA methylation and various metabolic, biochemical, and hematological parameters. In total, 80,728 participants from the Taiwan Biobank were enrolled for association analysis using functional MUC1 variants and a nearby gene regional plot association study. A subgroup of 1686 participants was recruited for MUC1 DNA methylation analysis. After Bonferroni correction, we found that two MUC1 variants, rs4072037 and rs12411216, were significantly associated with waist circumference, systolic blood pressure, hemoglobin A1C, renal functional parameters (blood urea nitrogen, serum creatinine levels, and estimated glomerular filtration rate), albuminuria, hematocrit, hemoglobin, red blood cell count, serum uric acid level, and gout risk, with both favorable and unfavorable effects. Causal inference analysis revealed that the association between the variants and gout was partially dependent on the serum uric acid level. Both gene variants showed genome-wide significant associations with MUC1 gene-body methylation. Regional plot association analysis further revealed lead single-nucleotide polymorphisms situated at the nearby TRIM46-MUC1-THBS3-MTX1 gene region for the studied phenotypes. In conclusion, our data demonstrated the pleiotropic effects of MUC1 variants with novel associations for gout, red blood cell parameters, and MUC1 DNA methylation. These results provide further evidence in understanding the critical role of TRIM46-MUC1-THBS3-MTX1 gene region variants in the pathogenesis of cardiometabolic, renal, and hematological disorders.
Collapse
|
40
|
5'-Nucleotidase Plays a Key Role in Uric Acid Metabolism of Bombyx mori. Cells 2021; 10:cells10092243. [PMID: 34571893 PMCID: PMC8468349 DOI: 10.3390/cells10092243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/06/2021] [Indexed: 01/26/2023] Open
Abstract
Uric acid (UA) is the end-product in the human purine metabolism pathway. The UA that accumulates in silkworm tissues is excreted as a nitrogen waste product. Here, we first validated that Bombyx mori has a homolog of the human gene that encodes the 5′-nucleotidase (5′N) involved in purine metabolism. The B. mori gene, Bm5′N, is located upstream of other genes involved in UA metabolism in the silkworm. Disruption of Bm5′N via the CRISPR/Cas9 system resulted in decreased UA levels in the silkworm epidermis and caused a translucent skin phenotype. When Bm5′N mutant silkworms were fed with the uric acid precursor inosine, the UA levels in the epidermis increased significantly. Furthermore, the metabolomic and transcriptomic analyses of Bm5′N mutants indicated that loss of the Bm5′N affected purine metabolism and the ABC transport pathway. Taken together, these results suggest that the UA pathway is conserved between the silkworm and humans and that the Bm5′N gene plays a crucial role in the uric acid metabolism of the silkworm. Thus, the silkworm may be a suitable model for the study of UA metabolism pathways relevant to human disease.
Collapse
|
41
|
Badii M, Gaal OI, Cleophas MC, Klück V, Davar R, Habibi E, Keating ST, Novakovic B, Helsen MM, Dalbeth N, Stamp LK, Macartney-Coxson D, Phipps-Green AJ, Stunnenberg HG, Dinarello CA, Merriman TR, Netea MG, Crişan TO, Joosten LAB. Urate-induced epigenetic modifications in myeloid cells. Arthritis Res Ther 2021; 23:202. [PMID: 34321071 PMCID: PMC8317351 DOI: 10.1186/s13075-021-02580-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES Hyperuricemia is a metabolic condition central to gout pathogenesis. Urate exposure primes human monocytes towards a higher capacity to produce and release IL-1β. In this study, we assessed the epigenetic processes associated to urate-mediated hyper-responsiveness. METHODS Freshly isolated human peripheral blood mononuclear cells or enriched monocytes were pre-treated with solubilized urate and stimulated with LPS with or without monosodium urate (MSU) crystals. Cytokine production was determined by ELISA. Histone epigenetic marks were assessed by sequencing immunoprecipitated chromatin. Mice were injected intraarticularly with MSU crystals and palmitate after inhibition of uricase and urate administration in the presence or absence of methylthioadenosine. DNA methylation was assessed by methylation array in whole blood of 76 participants with normouricemia or hyperuricemia. RESULTS High concentrations of urate enhanced the inflammatory response in vitro in human cells and in vivo in mice, and broad-spectrum methylation inhibitors reversed this effect. Assessment of histone 3 lysine 4 trimethylation (H3K4me3) and histone 3 lysine 27 acetylation (H3K27ac) revealed differences in urate-primed monocytes compared to controls. Differentially methylated regions (e.g. HLA-G, IFITM3, PRKAB2) were found in people with hyperuricemia compared to normouricemia in genes relevant for inflammatory cytokine signaling. CONCLUSION Urate alters the epigenetic landscape in selected human monocytes or whole blood of people with hyperuricemia compared to normouricemia. Both histone modifications and DNA methylation show differences depending on urate exposure. Subject to replication and validation, epigenetic changes in myeloid cells may be a therapeutic target in gout.
Collapse
Affiliation(s)
- M Badii
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
| | - O I Gaal
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
| | - M C Cleophas
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
| | - V Klück
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
| | - R Davar
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - E Habibi
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - S T Keating
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
| | - B Novakovic
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - M M Helsen
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - N Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - L K Stamp
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - D Macartney-Coxson
- Human Genomics, Institute of Environmental Science and Research (ESR), Wellington, New Zealand
| | - A J Phipps-Green
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - H G Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - C A Dinarello
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
- Department of Medicine, University of Colorado Denver, Aurora, CO, 80045, USA
| | - T R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
- Human Genomics Laboratory, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - T O Crişan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands
| | - L A B Joosten
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 8, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
42
|
Boocock J, Leask M, Okada Y, Matsuo H, Kawamura Y, Shi Y, Li C, Mount DB, Mandal AK, Wang W, Cadzow M, Gosling AL, Major TJ, Horsfield JA, Choi HK, Fadason T, O'Sullivan J, Stahl EA, Merriman TR. Genomic dissection of 43 serum urate-associated loci provides multiple insights into molecular mechanisms of urate control. Hum Mol Genet 2021; 29:923-943. [PMID: 31985003 DOI: 10.1093/hmg/ddaa013] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022] Open
Abstract
High serum urate is a prerequisite for gout and associated with metabolic disease. Genome-wide association studies (GWAS) have reported dozens of loci associated with serum urate control; however, there has been little progress in understanding the molecular basis of the associated loci. Here, we employed trans-ancestral meta-analysis using data from European and East Asian populations to identify 10 new loci for serum urate levels. Genome-wide colocalization with cis-expression quantitative trait loci (eQTL) identified a further five new candidate loci. By cis- and trans-eQTL colocalization analysis, we identified 34 and 20 genes, respectively, where the causal eQTL variant has a high likelihood that it is shared with the serum urate-associated locus. One new locus identified was SLC22A9 that encodes organic anion transporter 7 (OAT7). We demonstrate that OAT7 is a very weak urate-butyrate exchanger. Newly implicated genes identified in the eQTL analysis include those encoding proteins that make up the dystrophin complex, a scaffold for signaling proteins and transporters at the cell membrane; MLXIP that, with the previously identified MLXIPL, is a transcription factor that may regulate serum urate via the pentose-phosphate pathway and MRPS7 and IDH2 that encode proteins necessary for mitochondrial function. Functional fine mapping identified six loci (RREB1, INHBC, HLF, UBE2Q2, SFMBT1 and HNF4G) with colocalized eQTL containing putative causal SNPs. This systematic analysis of serum urate GWAS loci identified candidate causal genes at 24 loci and a network of previously unidentified genes likely involved in control of serum urate levels, further illuminating the molecular mechanisms of urate control.
Collapse
Affiliation(s)
- James Boocock
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Megan Leask
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan.,Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
| | | | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiaric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Changgui Li
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - David B Mount
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston MA, USA.,Renal Division, VA Boston Healthcare System, Harvard Medical School, Boston MA, USA
| | - Asim K Mandal
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston MA, USA
| | - Weiqing Wang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, New York, NY, USA
| | - Murray Cadzow
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Anna L Gosling
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Tanya J Major
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Julia A Horsfield
- Department of Pathology, Otago Medical School, University of Otago, Dunedin, New Zealand
| | - Hyon K Choi
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tayaza Fadason
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Eli A Stahl
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, New York, NY, USA
| | - Tony R Merriman
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
43
|
Zhu C, Sun B, Zhang B, Zhou Z. An update of genetics, co-morbidities and management of hyperuricaemia. Clin Exp Pharmacol Physiol 2021; 48:1305-1316. [PMID: 34133780 DOI: 10.1111/1440-1681.13539] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Hyperuricaemia (HU) caused by disorders of purine metabolism is a metabolic disease. A number of epidemiological reports have confirmed that HU is correlated with multiple disorders, such as chronic kidney diseases, cardiovascular disease and gout. Recent studies showed that the expression and functional changes of uric acid transporters, including URAT1, GLUT9 and ABCG2, were associated with HU. Moreover, a large number of genome-wide association studies have shown that these transporters' dysfunction leads to HU. In this review, we describe the recent progress of aetiology and related transporters of HU, and we also summarise the common co-morbidities possible mechanisms, as well as the potential pharmacological and non-pharmacological treatment methods for HU, aiming to provide new ideas for the treatment of HU.
Collapse
Affiliation(s)
- Chunsheng Zhu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bao Sun
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Bing Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Zheng Zhou
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
44
|
Nakatochi M, Toyoda Y, Kanai M, Nakayama A, Kawamura Y, Hishida A, Mikami H, Matsuo K, Takezaki T, Momozawa Y, Kamatani Y, Ichihara S, Shinomiya N, Yokota M, Wakai K, Okada Y, Matsuo H. An X chromosome-wide meta-analysis based on Japanese cohorts revealed that non-autosomal variations are associated with serum urate. Rheumatology (Oxford) 2021; 60:4430-4432. [PMID: 33944914 PMCID: PMC8409990 DOI: 10.1093/rheumatology/keab404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 02/04/2023] Open
Affiliation(s)
- Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Masahiro Kanai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruo Mikami
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshiro Takezaki
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | | | - Kenji Wakai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.,Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | | |
Collapse
|
45
|
Adaptive and maladaptive roles for ChREBP in the liver and pancreatic islets. J Biol Chem 2021; 296:100623. [PMID: 33812993 PMCID: PMC8102921 DOI: 10.1016/j.jbc.2021.100623] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive sugar consumption is a contributor to the worldwide epidemic of cardiometabolic disease. Understanding mechanisms by which sugar is sensed and regulates metabolic processes may provide new opportunities to prevent and treat these epidemics. Carbohydrate Responsive-Element Binding Protein (ChREBP) is a sugar-sensing transcription factor that mediates genomic responses to changes in carbohydrate abundance in key metabolic tissues. Carbohydrate metabolites activate the canonical form of ChREBP, ChREBP-alpha, which stimulates production of a potent, constitutively active ChREBP isoform called ChREBP-beta. Carbohydrate metabolites and other metabolic signals may also regulate ChREBP activity via posttranslational modifications including phosphorylation, acetylation, and O-GlcNAcylation that can affect ChREBP’s cellular localization, stability, binding to cofactors, and transcriptional activity. In this review, we discuss mechanisms regulating ChREBP activity and highlight phenotypes and controversies in ChREBP gain- and loss-of-function genetic rodent models focused on the liver and pancreatic islets.
Collapse
|
46
|
Toyoda Y, Kawamura Y, Nakayama A, Nakaoka H, Higashino T, Shimizu S, Ooyama H, Morimoto K, Uchida N, Shigesawa R, Takeuchi K, Inoue I, Ichida K, Suzuki H, Shinomiya N, Takada T, Matsuo H. Substantial anti-gout effect conferred by common and rare dysfunctional variants of URAT1/SLC22A12. Rheumatology (Oxford) 2021; 60:5224-5232. [PMID: 33821957 PMCID: PMC8566256 DOI: 10.1093/rheumatology/keab327] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/28/2021] [Indexed: 01/17/2023] Open
Abstract
Objectives Gout, caused by chronic elevation of serum uric acid levels, is the commonest form of inflammatory arthritis. The causative effect of common and rare variants of ATP-binding cassette transporter G2 (ABCG2/BCRP) on gout risk has been studied, but little attention has been paid to the effect of common (rs121907892, p.W258X) and rare variants of urate transporter 1 (URAT1/SLC22A12) on gout, despite dysfunctional variants of URAT1 having been identified as pathophysiological causes of renal hypouricaemia. Methods To address this important but overlooked issue, we investigated the effects of these URAT1 variants on gout susceptibility, using targeted exon sequencing on 480 clinically defined gout cases and 480 controls of Japanese males in combination with a series of functional analyses of newly identified URAT1 variants. Results Our results show that both common and rare dysfunctional variants of URAT1 markedly decrease the risk of gout (OR 0.0338, reciprocal OR 29.6, P = 7.66 × 10−8). Interestingly, we also found that the URAT1-related protective effect on gout eclipsed the ABCG2-related causative effect (OR 2.30–3.32). Our findings reveal only one dysfunctional variant of URAT1 to have a substantial anti-gout effect, even in the presence of causative variants of ABCG2, a ‘gout gene’. Conclusion Our findings provide a better understanding of gout/hyperuricaemia and its aetiology that is highly relevant to personalized health care. The substantial anti-gout effect of common and rare variants of URAT1 identified in the present study support the genetic concept of a ‘Common Disease, Multiple Common and Rare Variant’ model.
Collapse
Affiliation(s)
- Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan.,Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hirofumi Nakaoka
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan.,Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Toshihide Higashino
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | | | - Keito Morimoto
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Naohiro Uchida
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | | | - Kenji Takeuchi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Ituro Inoue
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Kimiyoshi Ichida
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.,Division of Kidney and Hypertension, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
47
|
Kawaguchi M, Nakayama A, Aoyagi Y, Nakamura T, Shimizu S, Kawamura Y, Takao M, Tamura T, Hishida A, Nagayoshi M, Nagase M, Ooyama K, Ooyama H, Shinomiya N, Matsuo H. Both variants of A1CF and BAZ1B genes are associated with gout susceptibility: a replication study and meta-analysis in a Japanese population. Hum Cell 2021; 34:293-299. [PMID: 33517564 PMCID: PMC7900071 DOI: 10.1007/s13577-021-00485-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/01/2021] [Indexed: 12/20/2022]
Abstract
Gout is a common type of acute arthritis that results from elevated serum uric acid (SUA) levels. Recent genome-wide association studies (GWASs) have revealed several novel single nucleotide polymorphism (SNPs) associated with SUA levels. Of these, rs10821905 of A1CF and rs1178977 of BAZ1B showed the greatest and the second greatest significant effect size for increasing SUA level in the Japanese population, but their association with gout is not clear. We examined their association with gout using 1411 clinically-defined Japanese gout patients and 1285 controls, and meta-analyzed our previous gout GWAS data to investigate any association with gout. Replication studies revealed both SNPs to be significantly associated with gout (P = 0.0366, odds ratio [OR] with 95% confidence interval [CI]: 1.30 [1.02-1.68] for rs10821905 of A1CF, P = 6.49 × 10-3, OR with 95% CI: 1.29 [1.07-1.55] for rs1178977 of BAZ1B). Meta-analysis also revealed a significant association with gout in both SNPs (Pmeta = 3.16 × 10-4, OR with 95% CI: 1.39 [1.17-1.66] for rs10821905 of A1CF, Pmeta = 7.28 × 10-5, OR with 95% CI 1.32 [1.15-1.51] for rs1178977 of BAZ1B). This study shows the first known association between SNPs of A1CF, BAZ1B and clinically-defined gout cases in Japanese. Our results also suggest a shared physiological/pathophysiological background between several populations, including Japanese, for both SUA increase and gout susceptibility. Our findings will not only assist the elucidation of the pathophysiology of gout and hyperuricemia, but also suggest new molecular targets.
Collapse
Affiliation(s)
- Makoto Kawaguchi
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yuka Aoyagi
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takahiro Nakamura
- Laboratory for Mathematics, Premedical Course, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Mikiya Takao
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Takashi Tamura
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mako Nagayoshi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | | | | | | | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| |
Collapse
|
48
|
Sinnott-Armstrong N, Naqvi S, Rivas M, Pritchard JK. GWAS of three molecular traits highlights core genes and pathways alongside a highly polygenic background. eLife 2021; 10:e58615. [PMID: 33587031 PMCID: PMC7884075 DOI: 10.7554/elife.58615] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 01/18/2021] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies (GWAS) have been used to study the genetic basis of a wide variety of complex diseases and other traits. We describe UK Biobank GWAS results for three molecular traits-urate, IGF-1, and testosterone-with better-understood biology than most other complex traits. We find that many of the most significant hits are readily interpretable. We observe huge enrichment of associations near genes involved in the relevant biosynthesis, transport, or signaling pathways. We show how GWAS data illuminate the biology of each trait, including differences in testosterone regulation between females and males. At the same time, even these molecular traits are highly polygenic, with many thousands of variants spread across the genome contributing to trait variance. In summary, for these three molecular traits we identify strong enrichment of signal in putative core gene sets, even while most of the SNP-based heritability is driven by a massively polygenic background.
Collapse
Affiliation(s)
| | - Sahin Naqvi
- Department of Genetics, Stanford UniversityStanfordUnited States
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Manuel Rivas
- Department of Biomedical Data Sciences, Stanford UniversityStanfordUnited States
| | - Jonathan K Pritchard
- Department of Genetics, Stanford UniversityStanfordUnited States
- Department of Biology, Stanford UniversityStanfordUnited States
| |
Collapse
|
49
|
Kawamura Y, Toyoda Y, Ohnishi T, Hisatomi R, Higashino T, Nakayama A, Shimizu S, Yanagi M, Kamimaki I, Fujimaru R, Suzuki H, Shinomiya N, Takada T, Matsuo H. Identification of a dysfunctional splicing mutation in the SLC22A12/URAT1 gene causing renal hypouricaemia type 1: a report on two families. Rheumatology (Oxford) 2021; 59:3988-3990. [PMID: 33011794 PMCID: PMC7733723 DOI: 10.1093/rheumatology/keaa461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa
| | - Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo
| | - Takuma Ohnishi
- Department of Pediatrics, National Hospital Organization Saitama Hospital, Wako
| | - Ryutaro Hisatomi
- Department of Pediatrics, Osaka City General Hospital, Osaka, Japan
| | - Toshihide Higashino
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa
| | - Masato Yanagi
- Department of Pediatrics, National Hospital Organization Saitama Hospital, Wako
| | - Isamu Kamimaki
- Department of Pediatrics, National Hospital Organization Saitama Hospital, Wako
| | - Rika Fujimaru
- Department of Pediatrics, Osaka City General Hospital, Osaka, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa
| |
Collapse
|
50
|
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: 0.5] [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
|