1
|
Liu T, Wang L, Ji L, Mu L, Wang K, Xu G, Wang S, Ma Q. Plantaginis Semen Ameliorates Hyperuricemia Induced by Potassium Oxonate. Int J Mol Sci 2024; 25:8548. [PMID: 39126116 PMCID: PMC11313179 DOI: 10.3390/ijms25158548] [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: 06/17/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Plantaginis semen is the dried ripe seed of Plantago asiatica L. or Plantago depressa Willd., which has a long history in alleviating hyperuricemia (HUA) and chronic kidney diseases. While the major chemical ingredients and mechanism remained to be illustrated. Therefore, this work aimed to elucidate the chemicals and working mechanisms of PS for HUA. UPLC-QE-Orbitrap-MS was applied to identify the main components of PS in vitro and in vivo. RNA sequencing (RNA-seq) was conducted to explore the gene expression profile, and the genes involved were further confirmed by real-time quantitative PCR (RT-qPCR). A total of 39 components were identified from PS, and 13 of them were detected in the rat serum after treating the rat with PS. The kidney tissue injury and serum uric acid (UA), xanthine oxidase (XOD), and cytokine levels were reversed by PS. Meanwhile, renal urate anion transporter 1 (Urat1) and glucose transporter 9 (Glut9) levels were reversed with PS treatment. RNA-seq analysis showed that the PPAR signaling pathway; glycine, serine, and threonine metabolism signaling pathway; and fatty acid metabolism signaling pathway were significantly modified by PS treatment. Further, the gene expression of Slc7a8, Pck1, Mgll, and Bhmt were significantly elevated, and Fkbp5 was downregulated, consistent with RNA-seq results. The PPAR signaling pathway involved Pparα, Pparγ, Lpl, Plin5, Atgl, and Hsl were elevated by PS treatment. URAT1 and PPARα proteins levels were confirmed by Western blotting. In conclusion, this study elucidates the chemical profile and working mechanisms of PS for prevention and therapy of HUA and provides a promising traditional Chinese medicine agency for HUA prophylaxis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shifeng Wang
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; (T.L.); (L.W.); (L.J.); (L.M.); (K.W.); (G.X.)
| | - Qun Ma
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; (T.L.); (L.W.); (L.J.); (L.M.); (K.W.); (G.X.)
| |
Collapse
|
2
|
Cho C, Kim B, Kim DS, Hwang MY, Shim I, Song M, Lee YC, Jung SH, Cho SK, Park WY, Myung W, Kim BJ, Do R, Choi HK, Merriman TR, Kim YJ, Won HH. Large-scale cross-ancestry genome-wide meta-analysis of serum urate. Nat Commun 2024; 15:3441. [PMID: 38658550 PMCID: PMC11043400 DOI: 10.1038/s41467-024-47805-4] [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: 08/08/2023] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Hyperuricemia is an essential causal risk factor for gout and is associated with cardiometabolic diseases. Given the limited contribution of East Asian ancestry to genome-wide association studies of serum urate, the genetic architecture of serum urate requires exploration. A large-scale cross-ancestry genome-wide association meta-analysis of 1,029,323 individuals and ancestry-specific meta-analysis identifies a total of 351 loci, including 17 previously unreported loci. The genetic architecture of serum urate control is similar between European and East Asian populations. A transcriptome-wide association study, enrichment analysis, and colocalization analysis in relevant tissues identify candidate serum urate-associated genes, including CTBP1, SKIV2L, and WWP2. A phenome-wide association study using polygenic risk scores identifies serum urate-correlated diseases including heart failure and hypertension. Mendelian randomization and mediation analyses show that serum urate-associated genes might have a causal relationship with serum urate-correlated diseases via mediation effects. This study elucidates our understanding of the genetic architecture of serum urate control.
Collapse
Affiliation(s)
- Chamlee Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Beomsu Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Dan Say Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Mi Yeong Hwang
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Injeong Shim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Minku Song
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea
| | - Yeong Chan Lee
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sung Kweon Cho
- Department of Pharmacology, Ajou University School of Medicine (AUSOM), Suwon, Republic of Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Bong-Jo Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hyon K Choi
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tony R Merriman
- Biochemistry Department, University of Otago, Dunedin, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Young Jin Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, Republic of Korea.
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Liu Y, Liu X, Wang M, Chen C, Li X, Liang Z, Shan Y, Yin Y, Sun F, Li Z, Li H. Characterizations of microRNAs involved in the molecular mechanisms underlying the therapeutic effects of noni ( Morinda citrifolia L.) fruit juice on hyperuricemia in mice. Front Nutr 2023; 10:1121734. [PMID: 37426193 PMCID: PMC10324520 DOI: 10.3389/fnut.2023.1121734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/06/2023] [Indexed: 07/11/2023] Open
Abstract
Background Hyperuricemia is generally defined as the high level of serum uric acid and is well known as an important risk factor for the development of various medical disorders. However, the medicinal treatment of hyperuricemia is frequently associated with multiple side-effects. Methods The therapeutic effect of noni (Morinda citrifolia L.) fruit juice on hyperuricemia and the underlying molecular mechanisms were investigated in mouse model of hyperuricemia induced by potassium oxonate using biochemical and high-throughput RNA sequencing analyses. Results The levels of serum uric acid (UA) and xanthine oxidase (XOD) in mice treated with noni fruit juice were significantly decreased, suggesting that the noni fruit juice could alleviate hyperuricemia by inhibiting the XOD activity and reducing the level of serum UA. The contents of both serum creatinine and blood urine nitrogen of the noni fruit juice group were significantly lower than those of the model group, suggesting that noni fruit juice promoted the excretion of UA without causing deleterious effect on the renal functions in mice. The differentially expressed microRNAs involved in the pathogenesis of hyperuricemia in mice were identified by RNA sequencing with their target genes further annotated based on both Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases to explore the metabolic pathways and molecular mechanisms underlying the therapeutic effect on hyperuricemia by noni fruit juice. Conclusion Our study provided strong experimental evidence to support the further investigations of the potential application of noni fruit juice in the treatment of hyperuricemia.
Collapse
Affiliation(s)
- Yue Liu
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Xianjun Liu
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Mengyuan Wang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Changwu Chen
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Xiaohong Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Zhiyong Liang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
- Qingdao Haoda Marine Biotechnology Co., Ltd., Qingdao, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Yuhe Yin
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, China
| | - Fengjie Sun
- School of Science and Technology, Georgia Gwinnett College, Lawrenceville, GA, United States
| | - Zhandong Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Hao Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| |
Collapse
|
4
|
Pack AI, Keenan BT. Rare Genetic Variants Provide Protection for Obstructive Sleep Apnea. Am J Respir Crit Care Med 2022; 206:1199-1200. [PMID: 35904813 PMCID: PMC9746843 DOI: 10.1164/rccm.202207-1414ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Allan I Pack
- Perelman School of Medicine University of Pennsylvania Philadelphia, Pennsylvania
| | - Brendan T Keenan
- Perelman School of Medicine University of Pennsylvania Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Li Y, Luo Y, Wang Q, Liu X. Detection and Quantification of the Relationship between the Ratio of Triglycerides over High-Density Lipoprotein Cholesterol and the Level of Serum Uric Acid: One Cross-Sectional Study. Int J Endocrinol 2022; 2022:1673335. [PMID: 39263260 PMCID: PMC11390236 DOI: 10.1155/2022/1673335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/25/2022] [Accepted: 08/16/2022] [Indexed: 09/13/2024] Open
Abstract
Background Hyperuricemia acts as an independently known risk factor for diabetes, cardiovascular disease, and gout. It was previously reported that the ratio of triglycerides to high-density lipoprotein cholesterol (TG/HDL-C) is not only an important marker of cardiovascular disease, stroke, atherosclerosis, and insulin resistance but is also associated with an elevated level of serum uric acid. However, it is still poorly understood what the association is between TG/HDL-C and serum uric acid levels. Hence, the aim of this research was to determine this association. Methods A total of 5,402 participants who underwent physical examinations in 2021 were analyzed during our cross-sectional research. In order to verify this correlation between TG/HDL-C and uric acid, we performed both a generalized additive model (GAM) and a smoothing curve fit. We also performed receiver operating characteristic (ROC) curves for evaluation of differences in clinical risk factor models in identifying hyperuricemia risk before and after the introduction of TG/HDL-C. Results Upon adjustment for confounders, we found that there was a nonlinear positive correlation between TG/HDL-C and the level of uric acid, and the inflection point was 1.41. When TG/HDL-C was less than 1.41, the effect size was 40.56 (19.08-62.04, P = 0.0002), whereas when TG/HDL-C was more than 1.41 the effect size was 17.18 (3.70-30.65, P=0.0125). As shown by the ROC curve, a significant increase in the area under the curve (AUC) was observed upon the introduction of TG/HDL-C into the established risk factor model which elevated from 0.7206(0.7053-0.7359, P < 0.05) to 0.8291 (0.8175-0.8407, P < 0.05). Conclusion Therefore, TG/HDL-C is positively and nonlinearly correlated to the level of uric acid, and the inflection point is 1.41. Furthermore, TG/HDL-C leads to an improvement in hyperuricemia risk stratification.
Collapse
Affiliation(s)
- Yuexi Li
- Health Management Center, Deyang People's Hospital, No. 173, Taishan North Road, Deyang, Sichuan, China
| | - Yuhan Luo
- Health Management Center, Deyang People's Hospital, No. 173, Taishan North Road, Deyang, Sichuan, China
| | - Qiaoli Wang
- Health Management Center, Deyang People's Hospital, No. 173, Taishan North Road, Deyang, Sichuan, China
| | - Xiaoqin Liu
- Health Management Center, Deyang People's Hospital, No. 173, Taishan North Road, Deyang, Sichuan, China
| |
Collapse
|
6
|
Misawa K. Genotype Value Decomposition: Simple Methods for the Computation of Kernel Statistics. ADVANCED GENETICS (HOBOKEN, N.J.) 2022; 3:2100066. [PMID: 36620199 PMCID: PMC9744480 DOI: 10.1002/ggn2.202100066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Indexed: 01/11/2023]
Abstract
Recent advances in sequencing technologies enable genome-wide analyses for thousands of individuals. The sequential kernel association test (SKAT) is a widely used method to test for associations between a phenotype and a set of rare variants. As the sample size of human genetics studies increases, the computational time required to calculate a kernel is becoming more and more problematic. In this study, a new method to obtain kernel statistics without calculating a kernel matrix is proposed. A simple method for the computation of two kernel statistics, namely, a kernel statistic based on a genetic relationship matrix (GRM) and one based on an identity by state (IBS) matrix, are proposed. By using this method, calculation of the kernel statistics can be conducted using vector calculation without matrix calculation. The proposed method enables one to conduct SKAT for large samples of human genetics.
Collapse
Affiliation(s)
- Kazuharu Misawa
- Department of Human GeneticsYokohama City University Graduate School of Medicine3‐9 Fukuura, Kanazawa‐kuYokohama236‐0004Japan
| |
Collapse
|
7
|
Nian YL, You CG. Susceptibility genes of hyperuricemia and gout. Hereditas 2022; 159:30. [PMID: 35922835 PMCID: PMC9351246 DOI: 10.1186/s41065-022-00243-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/03/2022] [Indexed: 11/10/2022] Open
Abstract
Gout is a chronic metabolic disease that seriously affects human health. It is also a major challenge facing the world, which has brought a heavy burden to patients and society. Hyperuricemia (HUA) is the most important risk factor for gout. In recent years, with the improvement of living standards and the change of dietary habits, the incidence of gout in the world has increased dramatically, and gradually tends to be younger. An increasing number of studies have shown that gene mutations may play an important role in the development of HUA and gout. Therefore, we reviewed the existing literature and summarized the susceptibility genes and research status of HUA and gout, in order to provide reference for the early diagnosis, individualized treatment and the development of new targeted drugs of HUA and gout.
Collapse
Affiliation(s)
- Yue-Li Nian
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Chong-Ge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
8
|
Liu P, Fang M, Luo Y, Zheng F, Jin Y, Cheng F, Zhu H, Jin X. Rare Variants in Inborn Errors of Immunity Genes Associated With Covid-19 Severity. Front Cell Infect Microbiol 2022; 12:888582. [PMID: 35694544 PMCID: PMC9184678 DOI: 10.3389/fcimb.2022.888582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/21/2022] [Indexed: 01/08/2023] Open
Abstract
Host genetic factors have been shown to play an important role in SARS-CoV-2 infection and the course of Covid-19 disease. The genetic contributions of common variants influencing Covid-19 susceptibility and severity have been extensively studied in diverse populations. However, the studies of rare genetic defects arising from inborn errors of immunity (IEI) are relatively few, especially in the Chinese population. To fill this gap, we used a deeply sequenced dataset of nearly 500 patients, all of Chinese descent, to investigate putative functional rare variants. Specifically, we annotated rare variants in our call set and selected likely deleterious missense (LDM) and high-confidence predicted loss-of-function (HC-pLoF) variants. Further, we analyzed LDM and HC-pLoF variants between non-severe and severe Covid-19 patients by (a) performing gene- and pathway-level association analyses, (b) testing the number of mutations in previously reported genes mapped from LDM and HC-pLoF variants, and (c) uncovering candidate genes via protein-protein interaction (PPI) network analysis of Covid-19-related genes and genes defined from LDM and HC-pLoF variants. From our analyses, we found that (a) pathways Tuberculosis (hsa:05152), Primary Immunodeficiency (hsa:05340), and Influenza A (hsa:05164) showed significant enrichment in severe patients compared to the non-severe ones, (b) HC-pLoF mutations were enriched in Covid-19-related genes in severe patients, and (c) several candidate genes, such as IL12RB1, TBK1, TLR3, and IFNGR2, are uncovered by PPI network analysis and worth further investigation. These regions generally play an essential role in regulating antiviral innate immunity responses to foreign pathogens and in responding to many inflammatory diseases. We believe that our identified candidate genes/pathways can be potentially used as Covid-19 diagnostic markers and help distinguish patients at higher risk.
Collapse
Affiliation(s)
- Panhong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Mingyan Fang
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
- Beijing Genomeics Institute In Singapore, BGI-Singapore, Singapore, Singapore
| | - Yuxue Luo
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Fang Zheng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fanjun Cheng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huanhuan Zhu
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
- *Correspondence: Xin Jin, ; Huanhuan Zhu,
| | - Xin Jin
- Beijing Genomeics Institute At Shenzhen, BGI-Shenzhen, Shenzhen, China
- Beijing Genomeics Institute In Singapore, BGI-Singapore, Singapore, Singapore
- School of Medicine, South China University of Technology, Guangzhou, China
- *Correspondence: Xin Jin, ; Huanhuan Zhu,
| |
Collapse
|
9
|
Otani N, Ouchi M, Misawa K, Hisatome I, Anzai N. Hypouricemia and Urate Transporters. Biomedicines 2022; 10:biomedicines10030652. [PMID: 35327453 PMCID: PMC8945357 DOI: 10.3390/biomedicines10030652] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/07/2023] Open
Abstract
Hypouricemia is recognized as a rare disorder, defined as a serum uric acid level of 2.0 mg/dL or less. Hypouricemia is divided into an overexcretion type and an underproduction type. The former typical disease is xanthinuria, and the latter is renal hypouricemia (RHUC). The frequency of nephrogenic hypouricemia due to a deficiency of URAT1 is high in Japan, accounting for most asymptomatic and persistent cases of hypouricemia. RHUC results in a high risk of exercise-induced acute kidney injury and urolithiasis. It is vital to promote research on RHUC, as this will lead not only to the elucidation of its pathophysiology but also to the development of new treatments for gout and hyperuricemia.
Collapse
Affiliation(s)
- Naoyuki Otani
- Department of Clinical Pharmacology and Therapeutics, Faculty of Medicine, Oita University, Yufu 879-5593, Oita, Japan;
| | - Motoshi Ouchi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Tochigi, Japan;
| | - Kazuharu Misawa
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Kanagawa, Japan;
| | - Ichiro Hisatome
- Yonago Medical Center, National Hospital Organization, Yonago 683-0006, Tottori, Japan;
- Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Sciences, Tottori University, Yonago 680-8550, Tottori, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Chiba, Japan
- Correspondence:
| |
Collapse
|
10
|
Méndez-Salazar EO, Martínez-Nava GA. Uric acid extrarenal excretion: the gut microbiome as an evident yet understated factor in gout development. Rheumatol Int 2021; 42:403-412. [PMID: 34586473 DOI: 10.1007/s00296-021-05007-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/19/2021] [Indexed: 12/19/2022]
Abstract
Humans do not produce uricase, an enzyme responsible for degrading uric acid. However, some bacteria residing in the gut can degrade one-third of the dietary and endogenous uric acid generated daily. New insights based on metagenomic and metabolomic approaches provide a new interest in exploring the involvement of gut microbiota in gout. Nevertheless, the exact mechanisms underlying this association are complex and have not been widely discussed. In this study, we aimed to review the evidence that suggests uric acid extrarenal excretion and gut microbiome are potential risk factors for developing gout. A literature search was performed in PubMed, Web of Science, and Google Scholar using several keywords, including "gut microbiome AND gout". A remarkable intestinal dysbiosis and shifts in abundance of certain bacterial taxa in gout patients have been consistently reported among different studies. Under this condition, bacteria might have developed adaptive mechanisms for de novo biosynthesis and salvage of purines, and thus, a concomitant alteration in uric acid metabolism. Moreover, gut microbiota can produce substrates that might cross the portal vein so the liver can generate de novo purinogenic amino acids, as well as uric acid. Therefore, the extrarenal excretion of uric acid needs to be considered as a factor in gout development. Nevertheless, further studies are needed to fully understand the role of gut microbiome in uric acid production and its extrarenal excretion, and to point out possible bacteria or bacterial enzymes that could be used as probiotic coadjutant treatment in gout patients.
Collapse
Affiliation(s)
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico.
| |
Collapse
|
11
|
Sun HL, Wu YW, Bian HG, Yang H, Wang H, Meng XM, Jin J. Function of Uric Acid Transporters and Their Inhibitors in Hyperuricaemia. Front Pharmacol 2021; 12:667753. [PMID: 34335246 PMCID: PMC8317579 DOI: 10.3389/fphar.2021.667753] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Disorders of uric acid metabolism may be associated with pathological processes in many diseases, including diabetes mellitus, cardiovascular disease, and kidney disease. These diseases can further promote uric acid accumulation in the body, leading to a vicious cycle. Preliminary studies have proven many mechanisms such as oxidative stress, lipid metabolism disorders, and rennin angiotensin axis involving in the progression of hyperuricaemia-related diseases. However, there is still lack of effective clinical treatment for hyperuricaemia. According to previous research results, NPT1, NPT4, OAT1, OAT2, OAT3, OAT4, URAT1, GLUT9, ABCG2, PDZK1, these urate transports are closely related to serum uric acid level. Targeting at urate transporters and urate-lowering drugs can enhance our understanding of hyperuricaemia and hyperuricaemia-related diseases. This review may put forward essential references or cross references to be contributed to further elucidate traditional and novel urate-lowering drugs benefits as well as provides theoretical support for the scientific research on hyperuricemia and related diseases.
Collapse
Affiliation(s)
- Hao-Lu Sun
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Yi-Wan Wu
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - He-Ge Bian
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Hui Yang
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Heng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Juan Jin
- Department of Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Pang H, Xia Y, Luo S, Huang G, Li X, Xie Z, Zhou Z. Emerging roles of rare and low-frequency genetic variants in type 1 diabetes mellitus. J Med Genet 2021; 58:289-296. [PMID: 33753534 PMCID: PMC8086251 DOI: 10.1136/jmedgenet-2020-107350] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is defined as an autoimmune disorder and has enormous complexity and heterogeneity. Although its precise pathogenic mechanisms are obscure, this disease is widely acknowledged to be precipitated by environmental factors in individuals with genetic susceptibility. To date, the known susceptibility loci, which have mostly been identified by genome-wide association studies, can explain 80%–85% of the heritability of T1DM. Researchers believe that at least a part of its missing genetic component is caused by undetected rare and low-frequency variants. Most common variants have only small to modest effect sizes, which increases the difficulty of dissecting their functions and restricts their potential clinical application. Intriguingly, many studies have indicated that rare and low-frequency variants have larger effect sizes and play more significant roles in susceptibility to common diseases, including T1DM, than common variants do. Therefore, better recognition of rare and low-frequency variants is beneficial for revealing the genetic architecture of T1DM and for providing new and potent therapeutic targets for this disease. Here, we will discuss existing challenges as well as the great significance of this field and review current knowledge of the contributions of rare and low-frequency variants to T1DM.
Collapse
Affiliation(s)
- Haipeng Pang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ying Xia
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuoming Luo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
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: 4] [Impact Index Per Article: 1.3] [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
|
14
|
Matsubayashi M, Sakaguchi YM, Sahara Y, Nanaura H, Kikuchi S, Asghari A, Bui L, Kobashigawa S, Nakanishi M, Nagata R, Matsui TK, Kashino G, Hasegawa M, Takasawa S, Eriguchi M, Tsuruya K, Nagamori S, Sugie K, Nakagawa T, Takasato M, Umetani M, Mori E. 27-Hydroxycholesterol regulates human SLC22A12 gene expression through estrogen receptor action. FASEB J 2020; 35:e21262. [PMID: 33368618 PMCID: PMC7771643 DOI: 10.1096/fj.202002077r] [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: 09/07/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
The excretion and reabsorption of uric acid both to and from urine are tightly regulated by uric acid transporters. Metabolic syndrome conditions, such as obesity, hypercholesterolemia, and insulin resistance, are believed to regulate the expression of uric acid transporters and decrease the excretion of uric acid. However, the mechanisms driving cholesterol impacts on uric acid transporters have been unknown. Here, we show that cholesterol metabolite 27‐hydroxycholesterol (27HC) upregulates the uric acid reabsorption transporter URAT1 encoded by SLC22A12 via estrogen receptors (ER). Transcriptional motif analysis showed that the SLC22A12 gene promoter has more estrogen response elements (EREs) than other uric acid reabsorption transporters such as SLC22A11 and SLC22A13, and 27HC‐activated SLC22A12 gene promoter via ER through EREs. Furthermore, 27HC increased SLC22A12 gene expression in human kidney organoids. Our results suggest that in hypercholesterolemic conditions, elevated levels of 27HC derived from cholesterol induce URAT1/SLC22A12 expression to increase uric acid reabsorption, and thereby, could increase serum uric acid levels.
Collapse
Affiliation(s)
| | | | - Yoshiki Sahara
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hitoki Nanaura
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan.,Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Sotaro Kikuchi
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan
| | - Arvand Asghari
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Linh Bui
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Shinko Kobashigawa
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan
| | - Mari Nakanishi
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan
| | - Riko Nagata
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan
| | - Takeshi K Matsui
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan.,Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Genro Kashino
- Radioisotope Research Center, Nara Medical University, Kashihara, Japan
| | - Masatoshi Hasegawa
- Department of Radiation Oncology, Nara Medical University, Kashihara, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara, Japan
| | | | - Kazuhiko Tsuruya
- Department of Nephrology, Nara Medical University, Kashihara, Japan
| | - Shushi Nagamori
- Department of Collaborative Research, Nara Medical University, Nara, Japan
| | - Kazuma Sugie
- Department of Neurology, Nara Medical University, Kashihara, Japan
| | - Takahiko Nakagawa
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan
| | - Minoru Takasato
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Michihisa Umetani
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.,HEALTH Research Institute, University of Houston, Houston, TX, USA
| | - Eiichiro Mori
- Department of Future Basic Medicine, Nara Medical University, Nara, Japan.,V-iCliniX Laboratory, Nara Medical University, Kashihara, Japan
| |
Collapse
|
15
|
Fukushima S, Farea M, Maeta K, Rani AQM, Fujioka K, Nishio H, Matsuo M. Dual Fluorescence Splicing Reporter Minigene Identifies an Antisense Oligonucleotide to Skip Exon v8 of the CD44 Gene. Int J Mol Sci 2020; 21:ijms21239136. [PMID: 33266296 PMCID: PMC7729581 DOI: 10.3390/ijms21239136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/25/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022] Open
Abstract
Splicing reporter minigenes are used in cell-based in vitro splicing studies. Exon skippable antisense oligonucleotide (ASO) has been identified using minigene splicing assays, but these assays include a time- and cost-consuming step of reverse transcription PCR amplification. To make in vitro splicing assay easier, a ready-made minigene (FMv2) amenable to quantitative splicing analysis by fluorescence microscopy was constructed. FMv2 was designed to encode two fluorescence proteins namely, mCherry, a transfection marker and split eGFP, a marker of splicing reaction. The split eGFP was intervened by an artificial intron containing a multicloning site sequence. Expectedly, FMv2 transfected HeLa cells produced not only red mCherry but also green eGFP signals. Transfection of FMv2CD44v8, a modified clone of FMv2 carrying an insertion of CD44 exon v8 in the multicloning site, that was applied to screen exon v8 skippable ASO, produced only red signals. Among seven different ASOs tested against exon v8, ASO#14 produced the highest index of green signal positive cells. Hence, ASO#14 was the most efficient exon v8 skippable ASO. Notably, the well containing ASO#14 was clearly identified among the 96 wells containing randomly added ASOs, enabling high throughput screening. A ready-made FMv2 is expected to contribute to identify exon skippable ASOs.
Collapse
Affiliation(s)
- Sachiyo Fukushima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (S.F.); (K.F.)
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe 651-2180, Japan; (M.F.); (K.M.); (A.Q.M.R.); (H.N.)
| | - Manal Farea
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe 651-2180, Japan; (M.F.); (K.M.); (A.Q.M.R.); (H.N.)
| | - Kazuhiro Maeta
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe 651-2180, Japan; (M.F.); (K.M.); (A.Q.M.R.); (H.N.)
- KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan
| | - Abdul Qawee Mahyoob Rani
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe 651-2180, Japan; (M.F.); (K.M.); (A.Q.M.R.); (H.N.)
- KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan
| | - Kazumichi Fujioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (S.F.); (K.F.)
| | - Hisahide Nishio
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe 651-2180, Japan; (M.F.); (K.M.); (A.Q.M.R.); (H.N.)
- Department of Occupational Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan
| | - Masafumi Matsuo
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe 651-2180, Japan; (M.F.); (K.M.); (A.Q.M.R.); (H.N.)
- KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan
- Correspondence: ; Tel.: +81-78-974-6194
| |
Collapse
|
16
|
Mishima E, Anzai N, Miyazaki M, Abe T. Uric Acid Elevation by Favipiravir, an Antiviral Drug. TOHOKU J EXP MED 2020; 251:87-90. [PMID: 32536670 DOI: 10.1620/tjem.251.87] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In light of the recent pandemic, favipiravir (Avigan®), a purine nucleic acid analog and antiviral agent approved for use in influenza in Japan, is being studied for the treatment of coronavirus disease 2019 (COVID-19). Increase in blood uric acid level is a frequent side effect of favipiravir. Here, we discussed the mechanism of blood uric acid elevation during favipiravir treatment. Favipiravir is metabolized to an inactive metabolite M1 by aldehyde oxidase and xanthine oxidase, and excreted into urine. In the kidney, uric acid handling is regulated by the balance of reabsorption and tubular secretion in the proximal tubules. Favipiravir and M1 act as moderate inhibitors of organic anion transporter 1 and 3 (OAT1 and OAT3), which are involved in uric acid excretion in the kidney. In addition, M1 enhances uric acid reuptake via urate transporter 1 (URAT1) in the renal proximal tubules. Thus, favipiravir is thought to decrease uric acid excretion into urine, resulting in elevation of uric acid levels in blood. Elevated uric acid levels were returned to normal after discontinuation of favipiravir, and favipiravir is not used for long periods of time for the treatment of viral infection. Thus, the effect on blood uric acid levels was subclinical in most studies. Nevertheless, the adverse effect of favipiravir might be clinically important in patients with a history of gout, hyperuricemia, kidney function impairment (in which blood concentration of M1 increases), and where there is concomitant use of other drugs affecting blood uric acid elevation.
Collapse
Affiliation(s)
- Eikan Mishima
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine
| | - Mariko Miyazaki
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine
| | - Takaaki Abe
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine.,Division of Medical Science, Tohoku University Graduate School of Biomedical Engineering.,Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine
| |
Collapse
|