1
|
Han R, Wang Z, Li Y, Ke L, Li X, Li C, Tian Z, Liu X. Gut microbiota Lactobacillus johnsonii alleviates hyperuricemia by modulating intestinal urate and gut microbiota-derived butyrate. Chin Med J (Engl) 2025:00029330-990000000-01534. [PMID: 40304365 DOI: 10.1097/cm9.0000000000003603] [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: 09/28/2024] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Gut microbiota are important for uric acid (UA) metabolism within hyperuricemia (HUA); however, the underlying mechanisms of how the gut microbiota regulate intestinal UA metabolism remain unclear. This study aimed to explore the function of the intestine in HUA and to further reveal the possible mechanism. METHODS We conducted gut microbiota depletion to validate the role of gut microbiota in UA metabolism. A mouse model of HUA was established, and the gut microbiota and microbiome-derived metabolites were analyzed via 16S RNA gene sequencing and metabolomics analysis. The mechanism of the gut microbiota in HUA was elucidated by in vivo and in vitro experiments. RESULTS Antibiotic treatment elevated serum UA, disturbed purine metabolism, and decreased the relative abundance of Lactobacillus. HUA mice had a lower relative abundance of Lactobacillus johnsonii (L. johnsonii) and decreased gut butyrate concentration. Supplementation of L. johnsonii significantly reduces serum UA in hyperuricemia mice by preventing UA synthesis and promoting the excretion of gut purine metabolites. In addition, L. johnsonii enhanced intestinal UA excretion by heightening the urate transporter ABCG2 (adenosine triphosphate-binding cassette transporter, subfamily G, member 2) expression, and increasing the levels of butyrate, which upregulated ABCG2 expression via the Wnt5a/b/β-catenin signaling pathway. CONCLUSION Our results suggest that gut microbiota and microbiota-derived metabolites directly regulate gut UA metabolism, highlighting potential applications in the treatment of diet-induced HUA by targeting gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Rongshuang Han
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Zan Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yukun Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Leyong Ke
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, China
| | - Xiang Li
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, China
| | - Changgui Li
- Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong 266003, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Xin Liu
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, China
| |
Collapse
|
2
|
Ye P, Liu W, Tang X, Liu M, Han J, Wang X, Zhu J, He X, Zhu X, Cao M, Zhao L, Ren Q. Effects of hydroxypropyl starch on intestinal health and transcriptome of geese. Sci Rep 2025; 15:12284. [PMID: 40210970 PMCID: PMC11986093 DOI: 10.1038/s41598-025-96020-8] [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: 11/09/2024] [Accepted: 03/25/2025] [Indexed: 04/12/2025] Open
Abstract
In recent years, gout resulting from uric acid metabolism disorders has led to significant economic losses in goose production. The intestine is a vital organ crucial for uric acid metabolism. Hydroxypropyl starch (HPS) is a resistant starch modified from natural starch, which can enhance intestinal health as a dietary ingredient fiber. In this study, 240 30-day-old Yangzhou geese with similar body weights were divided into three groups: The control group (CG) received a basal diet + 5% corn starch; the hydroxypropyl starch group (HPS) and the sodium urate group (SU) were given a basal diet + 5% hydroxypropyl starch. The experiment lasted for 21 days, and the SU group was administered 30 mg of sodium urate per day during the last 4 days of the study. The results indicated that the level of uric acid in the HPS group was 56.6 µmol/L, significantly lower than that in the CG group (70.8 µmol/L) and the SU group (129.7 µmol/L). The morphological findings revealed that the ileum of the CG group and the SU group exhibited varying degrees of damage, while the HPS group maintained complete structure. The villus height and the ratio of villus height to crypt depth in the HPS group were significantly higher compared to those in the CG and SU groups, while the crypt depth was significantly lower than that in the SU group. A total of 1462 differentially expressed genes (DEGs) were identified at the transcriptome level. GO and KEGG functional enrichment analyses indicated that the DEGs were significantly enriched in the Brush border membrane, Brush border, PPAR signaling pathway, PI3K-Akt signaling pathway, and other related processes. Subsequent analysis revealed that HPS up-regulated the expression of genes associated with intestinal function (such as SLC5A12 and SLC5A8), structure (including NR5A2, IPMK), and uric acid metabolism (PDZK1). The accuracy and reliability of transcriptome sequencing data were confirmed by RT-qPCR. In this study, we systematically demonstrated that HPS can improve intestinal morphology and reduce serum uric acid levels, emphasizing its potential as a dietary supplement for geese.
Collapse
Affiliation(s)
- Pengfei Ye
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China.
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China.
| | - Wenquan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xiaotong Tang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Mengxue Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Jingfan Han
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xiaoxue Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Jie Zhu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xiaorong He
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Xueqi Zhu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China
| | - Mixia Cao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Lei Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Qingchang Ren
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China.
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, Chuzhou, 233100, China.
| |
Collapse
|
3
|
Zhou S, Wen X, Lessing DJ, Chu W. Uric Acid-Degrading Lacticaseibacillus paracasei CPU202306 Ameliorates Hyperuricemia by Regulating Uric Acid Metabolism and Intestinal Microecology in Mice. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10532-3. [PMID: 40205164 DOI: 10.1007/s12602-025-10532-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Hyperuricemia, characterized by elevated levels of uric acid in the blood, poses a significant health threat due to its association with various adverse health outcomes, and lactic acid bacteria from the gut microbiota may offer solutions. Our investigation focused on Lacticaseibacillus paracasei CPU202306, isolated from fermented pickles for its potent uric acid degradation and probiotic properties. This bacterium effectively reduced blood uric acid levels by breaking down uric acid and inhibiting hepatic xanthine oxidase (XOD) and adenosine deaminase (ADA) enzymes. Additionally, it stimulated the production of short-chain fatty acid (SCFAs) in the colon, enhancing the expression of uric acid secretion transport proteins (ATP-binding cassette sub-family G member 2 and organic anion transporter 3) while suppressing absorption transport proteins (glucose transporter 9 and uric acid transporter 1). This orchestrated process promoted uric acid excretion. L. paracasei CPU202306 also improved gut microbiota health by reinforcing tight junction proteins, shifting the microbiota to a healthier composition, and reducing harmful bacteria. This transformation inhibited kidney TLR4/MyD88/NF-κB inflammatory signaling, leading to a significant decrease in pro-inflammatory cytokines and an increase in anti-inflammatory cytokines, mitigating kidney inflammation. Furthermore, the bacterium supported kidney health by influencing amino acid metabolic pathways linked to the gut-kidney axis. In summary, our study highlights the diverse mechanisms through which L. paracasei CPU202306 addresses hyperuricemia, showcasing its therapeutic potential for this condition.
Collapse
Affiliation(s)
- Shuxin Zhou
- Department of Microbiology and Synthetic Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin Wen
- Department of Microbiology and Synthetic Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Duncan James Lessing
- Department of Microbiology and Synthetic Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Weihua Chu
- Department of Microbiology and Synthetic Biology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
4
|
Wang T, Li L, Liu L, Tan R, Wu Q, Zhu X, Hua H, Dai Y, Li H, Mao J, Zhao J, Yin Z. Overview of pharmacodynamical research of traditional Chinese medicine on hyperuricemic nephropathy: from the perspective of dual-regulatory effect on the intestines and kidneys. Front Pharmacol 2025; 16:1517047. [PMID: 40264662 PMCID: PMC12011833 DOI: 10.3389/fphar.2025.1517047] [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: 10/25/2024] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
Uncontrolled hyperuricemia contributes to chronic kidney disease, characterized by renal inflammatory cell infiltration and tubulointerstitial fibrosis, eventually leading to renal failure. In addition to liver and kidney, the intestine tract plays a vital role in the development and progression of hyperuricemia and hyperuricemic nephropathy (HN) through various mechanisms. The conventional therapeutic strategy for HN is uric acid-lowering therapy (ULT) and renal protection; however, unsatisfactory results are often obtained in clinical practice. Growing evidence has demonstrated that traditional Chinese medicines (TCMs) achieve an anti-HN effect by modulating multiple targets and approaches with fewer side effects. Therefore, this paper reviews the pathogenesis of HN, including the role of soluble and insoluble urates in kidney and intestine, and the role of intestinal tract in the progression of HN. Meanwhile, the recent advancements in TCMs for the treatment of HN are summarized and analyzed, with a focus on their modulation of intestinal flora and metabolites, urate-related transporters, immuno-inflammation and barrier function in the intestines. Notably, for the first time, we propose the perspective that TCMs treat HN through a dual-regulatory effect on the intestines and kidneys. Additionally, the problems existing in current research and the feasible research strategies combined with emerging technologies such as fermentation and nanotechnology are discussed, thus providing novel ideas for HN management.
Collapse
Affiliation(s)
- Ting Wang
- Country School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Li Li
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Li Liu
- Sichuan Institute for Translational Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Ruirong Tan
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Qinxuan Wu
- Changsha Medical University, Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha, China
| | - Xin Zhu
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Hua Hua
- Sichuan Institute for Translational Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Ying Dai
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Huan Li
- Sichuan Acupuncture and Moxibustion School, Chengdu, China
| | - Jiuzhou Mao
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
| | - Junning Zhao
- Country School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Institute for Translational Chinese Medicine, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Zhujun Yin
- Sichuan Academy of Chinese Medicine Sciences, Institute of Pharmacology & Toxicology of Chinese Materia Medica, Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Engineering Research Center for Formation Principle and Quality Evaluation of Genuine Medicinal Materials in Sichuan Province, Chengdu, China
- Changsha Medical University, Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha, China
| |
Collapse
|
5
|
Chen Y, Yang H, Wang Z. A dynamic study on serum biomarkers and kidney injury in a gosling model of hyperuricemia. Poult Sci 2025; 104:105144. [PMID: 40209470 PMCID: PMC12008651 DOI: 10.1016/j.psj.2025.105144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/30/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025] Open
Abstract
Hyperuricemia, a metabolic disorder characterized by elevated serum uric acid levels, poses significant challenges to poultry health and productivity. This study aimed to establish a hyperuricemia model in goslings through a multi-factorial induction approach using a high-purine, high-protein, and high-calcium diet, to investigate its dynamic progression, serological markers, and renal pathology. A total of 160 male Sanhua goslings were randomly assigned to control and model groups and fed either a basal diet or a high-purine, high-protein, and high-calcium diet for 18 days. Key findings revealed significant elevations in serum uric acid, creatinine, and urea nitrogen levels in the model group from day 5 onward, peaking on day 13 (p < 0.01). Activities of xanthine oxidase and adenosine deaminase were consistently higher in the model group, indicating enhanced purine metabolism and oxidative stress. Histopathological analysis showed progressive renal damage, including tubular degeneration, interstitial fibrosis, and glomerular injury, becoming evident by day 7 and severe by day 13. Ultrastructural examination further revealed mitochondrial dysfunction, podocyte effacement, and basement membrane disruption in the model group. Behavioral abnormalities, such as reduced mobility and the "shrinking neck" phenomenon, were observed from day 9, reflecting systemic disease severity. The dynamic expression of fibrosis-related markers, including Col-1, Col-3, and α-SMA, demonstrated the progression of renal fibrosis and extracellular matrix remodeling. These findings identify biomarkers for hyperuricemia progression and suggest potential therapeutic targets. This study establishes a comprehensive framework for understanding hyperuricemia-associated renal pathology and lays the groundwork for developing strategies to mitigate its impact on poultry health and productivity.
Collapse
Affiliation(s)
- Yuanjing Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
| | - Haiming Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Zhiyue Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China; College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China.
| |
Collapse
|
6
|
Zhao J, Fu Y, Qiu H. Effect and mechanism of Plantaginis Semen polysaccharides on intestinal microecology in rats with hyperuricemia. Front Microbiol 2025; 16:1555734. [PMID: 40207160 PMCID: PMC11979160 DOI: 10.3389/fmicb.2025.1555734] [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: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Hyperuricemia (HUA) is characterized by metabolic abnormalities in purine metabolism, leading to an excessive accumulation of uric acid in the bloodstream. This condition is not only prevalent but also significantly linked to the exacerbation of various chronic diseases, including renal impairment. Notably, asymptomatic hyperuricemia is frequently associated with inflammatory responses and alterations in intestinal microbiota. Thus, it is imperative to explore effective therapeutic interventions for HUA to mitigate its associated health risks. Methods The present study aimed to elucidate the protective effects of Plantaginis Semen polysaccharides (PSP) in a rat model of hyperuricemia induced by adenine (AD) and potassium oxonate (PO) gavage. Over a treatment period of five weeks, the animals received either PSP or allopurinol (AL). Comprehensive assessments were performed, including blood biochemistry analysis, histopathological evaluation, Western blot analyses to investigate the expression levels of key renal transport proteins, as well as 16S rRNA sequencing to explore microbiota shifts. Results The findings demonstrated that PSP significantly decreased serum uric acid (UA) levels and alleviated renal dysfunction through modulation of xanthine oxidase (XOD) and adenosine deaminase (ADA) serum concentrations and the expression of renal transporters, namely glucose transporter protein 9 (GLUT9), urate transporter 1 (URAT1), ATP-binding cassette superfamily member 2 (ABCG2), and organic anion transporter 1 (OAT1). Furthermore, PSP exhibited notable anti-inflammatory properties, reflected in the reduced levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). Conclusion In summary, the present study substantiates the therapeutic potential of PSP in treating hyperuricemia through its dual action of lowering uric acid levels and imparting anti-inflammatory effects. The observed modulation of gut microbiota further supports the role of PSP in maintaining metabolic homeostasis. Future investigations should focus on the clinical applicability of PSP and elucidate the mechanisms underlying its beneficial impacts on hyperuricemia and associated metabolic disorders.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, China
- Department of Medical Laboratory, The First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yu Fu
- Laboratory Department, Center for Disease Control and Prevention of Jiamusi, Jiamusi, China
| | - Hongbin Qiu
- Key Laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
7
|
Wang H, Xi Y, Gu F, Peng L, Li J. Protective Effects of a Polyherbal Mixture on Intestinal Injury via the NF-κB Signaling Pathway and Gut Microbiota Modulation in Hyperuricemic Mice. Foods 2025; 14:1118. [PMID: 40238278 PMCID: PMC11988963 DOI: 10.3390/foods14071118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
This study investigated the protective effects of a polyherbal tea (PHT) on intestinal injury in hyperuricemia (HUA) mice and the underlying mechanisms. PHT was orally administered to mice for 49 days, while potassium oxonate and hypoxanthine were administered 7 days after PHT administration and continued for 42 days to cause HUA. Treatment with PHT significantly reduced serum uric acid and blood urea nitrogen levels in HUA mice. It also inhibited liver xanthine oxidase activity and promoted intestinal uric acid excretion through the upregulation of transporters GLUT9 and ABCG2. Intestinal barrier integrity was reinforced, as evidenced by the restoration of the villous structure, reduction in edema, and upregulation of tight junction proteins (occludin, ZO-1) and mucin (MUC2). Moreover, PHT suppressed serum LPS levels and inhibited the NF-κB pathway, leading to a reduction in TNF-α and IL-6 levels in the gut. Gut microbiota analysis revealed PHT reversed dysbiosis, enriching beneficial bacteria like Duncaniella sp. and Heminiphilus faecis. By UPLC-MS analysis, 154 compounds of PHT persisted in the gut, suggesting that these compounds are likely to modulate both intestinal barrier function and gut microbiota. These findings suggest that this PHT may have potential as a functional food for the prevention of hyperuricemia.
Collapse
Affiliation(s)
- Haoluan Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (H.W.); (F.G.); (L.P.)
- Key Laboratory of Green and Low-Carbon Processing Technology for Plant-Based Food of China National Light Industry Council, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Yu Xi
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (H.W.); (F.G.); (L.P.)
- Key Laboratory of Green and Low-Carbon Processing Technology for Plant-Based Food of China National Light Industry Council, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Fengju Gu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (H.W.); (F.G.); (L.P.)
- Key Laboratory of Green and Low-Carbon Processing Technology for Plant-Based Food of China National Light Industry Council, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Linlin Peng
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (H.W.); (F.G.); (L.P.)
- Key Laboratory of Green and Low-Carbon Processing Technology for Plant-Based Food of China National Light Industry Council, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jian Li
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (H.W.); (F.G.); (L.P.)
- Key Laboratory of Green and Low-Carbon Processing Technology for Plant-Based Food of China National Light Industry Council, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
8
|
Bond C, Bednarski OJ, Datta D, Namazzi R, Opoka RO, Lima-Cooper G, Batte A, Udumula K, Balasubramani D, Vasquez M, Rodriguez A, Liepmann C, Bangirana P, Abreu M, Schwantes-An TH, Zhao Y, El-Achkar TM, Schmidt NW, John CC, Conroy AL. Elevated uric acid levels, mortality and cognitive impairment in children with severe malaria. Nat Med 2025; 31:777-787. [PMID: 39856335 DOI: 10.1038/s41591-024-03430-8] [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: 04/08/2024] [Accepted: 11/26/2024] [Indexed: 01/27/2025]
Abstract
We investigated the role of uric acid in the pathogenesis of severe malaria (SM) in two independent cohorts of children with SM. Hyperuricemia (blood uric acid ≥ 7 mg dl-1) was present in 25% of children with SM and was associated with increased in-hospital mortality and postdischarge mortality in both cohorts. Increased blood uric acid levels were also associated with worse scores in overall cognition in children with SM < 5 years old in both cohorts. Hemolysis of infected red blood cells and impaired renal excretion of uric acid were the primary drivers of hyperuricemia in SM. Hyperuricemia was associated with multiple complications of SM, including acute kidney injury, acidosis, impaired perfusion, coma and intestinal injury with increases in the abundance of Gram-negative uricase-producing pathobionts (Escherichia and Shigella) in the stool. Clinical trials evaluating uric acid-lowering medications as adjunctive therapy for children with SM should be considered to improve survival and protect neurodevelopment.
Collapse
Affiliation(s)
- Caitlin Bond
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Olivia J Bednarski
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dibyadyuti Datta
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ruth Namazzi
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Robert O Opoka
- Aga Khan University East Africa Medical College, Nairobi, Kenya
| | - Giselle Lima-Cooper
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony Batte
- Child Health and Development Centre, Makerere University College of Health Sciences, Kampala, Uganda
| | - Keerthi Udumula
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Deepali Balasubramani
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marilyn Vasquez
- Department of Microbiology, New York University School of Medicine, New York City, NY, USA
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, New York City, NY, USA
| | - Claire Liepmann
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paul Bangirana
- Department of Psychiatry, Makerere University College of Health Sciences, Kampala, Uganda
| | - Marco Abreu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Zhao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tarek M El-Achkar
- Division of Nephrology, Indiana University and the Roudebush Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - Nathan W Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Andrea L Conroy
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
9
|
Li K, Xia X, Fu T, Ma Y, Wang Y, Fan M, Wang S, Xing G, Tong Y. Research progress on the mechanism of hyperuricemic nephropathy based on multi-omics technique: A review. Medicine (Baltimore) 2024; 103:e40975. [PMID: 39705438 PMCID: PMC11666175 DOI: 10.1097/md.0000000000040975] [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: 03/15/2024] [Revised: 07/24/2024] [Accepted: 11/27/2024] [Indexed: 12/22/2024] Open
Abstract
Hyperuricemic nephropathy is a metabolic disease in which renal uric acid deposition and excretion are impaired due to elevated levels of uric acid in the blood, leading to impaired renal tubule function and chronic renal disease. Hyperuricemic nephropathy is one of the important complications of hyperuricemia, which seriously affects the quality of life and prognosis of patients. The pathogenesis of hyperuricemic nephropathy involves a variety of factors, including: amino acid metabolism disorder, energy metabolism abnormality, increased nucleotide metabolism, lipid metabolism disorder and bile acid metabolism imbalance, REDOX process disorder, cell cycle and apoptosis imbalance, signal transduction and inflammatory response enhancement, and intestinal flora imbalance. In recent years, omics techniques such as metabolomics, transcriptomics and intestinal microecology have been used to reveal the metabolic, gene and microflora characteristics of hyperuricemic nephropathy from different levels, as well as their interactions and regulatory mechanisms. This paper reviews these studies, analyzes the existing problems and challenges, and puts forward future research directions and suggestions, aiming at providing new theoretical basis and practical guidance for the prevention and treatment of hyperuricemic nephropathy.
Collapse
Affiliation(s)
- Kaiqing Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xue Xia
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tong Fu
- Brandeis University, Waltham, China
| | - Yanchun Ma
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yingwei Wang
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Mingming Fan
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Songyan Wang
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Guoli Xing
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Tong
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
10
|
Wang Q, Liang J, Zou Q, Wang W, Yan G, Guo R, Yuan T, Wang Y, Liu X, Liu Z. Tryptophan Metabolism-Regulating Probiotics Alleviate Hyperuricemia by Protecting the Gut Barrier Integrity and Enhancing Colonic Uric Acid Excretion. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39564988 DOI: 10.1021/acs.jafc.4c07716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
The balance of gut microbiota affects uric acid synthesis and excretion, influencing the development of hyperuricemia. This study aimed to investigate the effects and mechanisms of probiotics on hyperuricemia and adenine- and potassium oxonate-induced colonic damage. After two months of gavage at 109 CFU/day, the probiotic strains Lactobacillus rhamnosus UA260 and Lactobacillus plantarum YU28, identified through in vitro screening, significantly reduced serum uric acid levels in hyperuricemia mice from 109.71 ± 56.33 to 38.76 ± 15.06 and 33.22 ± 6.91 μmol/L, respectively. These strains attenuated inflammatory, repaired gut barrier damage, and enhanced colonic uric acid transporter function, thereby promoting uric acid excretion. Furthermore, the probiotics significantly reshaped gut microbiota by increasing the abundance of beneficial bacteria, including Lactobacillus and Coprococcus, while modulating tryptophan, purine, and riboflavin metabolism. Changes in tryptophan metabolites, specifically indole-3-propionic acid and indole-3-acetic acid, were correlated with xanthine oxidase activity, colonic injury, and the expression of the uric acid transporter protein ABCG2 during treatment. Probiotics intervention activated aryl hydrocarbon receptor pathways. These findings suggest that probiotics alleviate hyperuricemia and colonic inflammatory by regulating gut microbiota composition and tryptophan microbial metabolite pathways. Probiotics that modulate tryptophan microbial metabolism may provide a potential strategy for treating or preventing hyperuricemia.
Collapse
Affiliation(s)
- Qianxu Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen 518000, China
| | - Jiarui Liang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen 518000, China
| | - Qianhui Zou
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen 518000, China
| | - Wenxiu Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Guiming Yan
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Rui Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen 518000, China
| | - Tian Yuan
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen 518000, China
| | - Yutang Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Zhigang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
- Northwest A&F University Shenzhen Research Institute, Shenzhen 518000, China
- Shaanxi Precision Nutrition and Health Research Institute, Xi'an 710300, China
| |
Collapse
|
11
|
Su Q, Li YC, Zhuang DH, Liu XY, Gao H, Li D, Chen Y, Ge MX, Han YM, Gao ZL, Yin FQ, Zhao L, Zhang YX, Yang LQ, Zhao Q, Luo YJ, Zhang Z, Kong QP. Rewiring of Uric Acid Metabolism in the Intestine Promotes High-Altitude Hypoxia Adaptation in Humans. Mol Biol Evol 2024; 41:msae233. [PMID: 39494496 DOI: 10.1093/molbev/msae233] [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: 05/28/2024] [Revised: 10/08/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024] Open
Abstract
Adaptation to high-altitude hypoxia is characterized by systemic and organ-specific metabolic changes. This study investigates whether intestinal metabolic rewiring is a contributing factor to hypoxia adaptation. We conducted a longitudinal analysis over 108 days, with seven time points, examining fecal metabolomic data from a cohort of 46 healthy male adults traveling from Chongqing (a.s.l. 243 m) to Lhasa (a.s.l. 3,658 m) and back. Our findings reveal that short-term hypoxia exposure significantly alters intestinal metabolic pathways, particularly those involving purines, pyrimidines, and amino acids. A notable observation was the significantly reduced level of intestinal uric acid, the end product of purine metabolism, during acclimatization (also called acclimation) and additional two long-term exposed cohorts (Han Chinese and Tibetans) residing in Shigatse, Xizang (a.s.l. 4,700 m), suggesting that low intestinal uric acid levels facilitate adaptation to high-altitude hypoxia. Integrative analyses with gut metagenomic data showed consistent trends in intestinal uric acid levels and the abundance of key uric acid-degrading bacteria, predominantly from the Lachnospiraceae family. The sustained high abundance of these bacteria in the long-term resident cohorts underscores their essential role in maintaining low intestinal uric acid levels. Collectively, these findings suggest that the rewiring of intestinal uric acid metabolism, potentially orchestrated by gut bacteria, is crucial for enhancing human resilience and adaptability in extreme environments.
Collapse
Affiliation(s)
- Qian Su
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu-Chun Li
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Dao-Hua Zhuang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Xin-Yuan Liu
- Department of Military Medical Geography, Army Health Service Training Base, Army Medical University, Chongqing 400038, China
| | - Han Gao
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Dong Li
- College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China
| | - Yu Chen
- Department of Military Medical Geography, Army Health Service Training Base, Army Medical University, Chongqing 400038, China
| | - Ming-Xia Ge
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Yi-Ming Han
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Zong-Liang Gao
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Fan-Qian Yin
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Long Zhao
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Yun-Xia Zhang
- The Second Affiliated Hospital, the School of Basic Medicine and Life Sciences, Hainan Medical University, Hainan 570102, China
| | - Li-Qin Yang
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Qin Zhao
- Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Yong-Jun Luo
- Department of Military Medical Geography, Army Health Service Training Base, Army Medical University, Chongqing 400038, China
| | - Zhigang Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, China
| | - Qing-Peng Kong
- Key Laboratory of Genetic Evolution & Animal Models (Chinese Academy of Sciences), Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| |
Collapse
|
12
|
Singh AK, Durairajan SSK, Iyaswamy A, Williams LL. Elucidating the role of gut microbiota dysbiosis in hyperuricemia and gout: Insights and therapeutic strategies. World J Gastroenterol 2024; 30:4404-4410. [PMID: 39494101 PMCID: PMC11525862 DOI: 10.3748/wjg.v30.i40.4404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/16/2024] Open
Abstract
Hyperuricemia (HUA) is a condition associated with a high concentration of uric acid (UA) in the bloodstream and can cause gout and chronic kidney disease. The gut microbiota of patients with gout and HUA is significantly altered compared to that of healthy people. This article focused on the complex interconnection between alterations in the gut microbiota and the development of this disorder. Some studies have suggested that changes in the composition, diversity, and activity of microbes play a key role in establishing and progressing HUA and gout pathogenesis. Therefore, we discussed how the gut microbiota contributes to HUA through purine metabolism, UA excretion, and intestinal inflammatory responses. We examined specific changes in the composition of the gut microbiota associated with gout and HUA, highlighting key bacterial taxa and the metabolic pathways involved. Additionally, we discussed the effect of conventional gout treatments on the gut microbiota composition, along with emerging therapeutic approaches that target the gut microbiome, such as the use of probiotics and prebiotics. We also provided insights into a study regarding the gut microbiota as a possible novel therapeutic intervention for gout treatment and dysbiosis-related diagnosis.
Collapse
Affiliation(s)
- Abhay Kumar Singh
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, TN 610005, India
| | - Siva Sundara Kumar Durairajan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, TN 610005, India
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ashok Iyaswamy
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Leonard L Williams
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| |
Collapse
|
13
|
Chen H, Yang G, Chen L, Zhao Y, Yao P, Li Y, Tang Y, Li D. Dietary polyunsaturated fatty acids intake is negatively associated with hyperuricemia: The National Health and Nutrition Examination Survey 2003-2015. Nutr Metab Cardiovasc Dis 2024; 34:2203-2216. [PMID: 39003131 DOI: 10.1016/j.numecd.2024.05.026] [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: 10/07/2023] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND AND AIMS The objective of this research was to explore the associations between dietary PUFAs intake and hyperuricemia risk. METHODS AND RESULTS Based on the National Health and Nutrition Examination Survey (NHANES) 2003-2015, all eligible individuals were divided into hyperuricemia and non-hyperuricemia groups based on diagnostic criteria for hyperuricemia (serum uric acid >420 μmol/L for men and >360 μmol/L for women). Multivariate-adjusted logistic regression was employed to explore the relationship between dietary PUFAs intake and hyperuricemia risk. Total PUFAs and their subtypes were modeled to isocalorically replace saturated fatty acids (SFAs) and monounsaturated fatty acids (MUFAs). Higher intake of n-3 PUFAs, n-6 PUFAs, linoleic acid (LA), alpha-linoleic acid (ALA), and non-marine PUFAs intake correlated with decreased hyperuricemia risk, with adjusted odds ratio (OR) and 95% confidence interval (95%CIs) were 0.77 (0.63, 0.93), 0.75 (0.61, 0.92), 0.75 (0.61, 0.91), 0.69 (0.55, 0.87), and 0.73 (0.59, 0.91), respectively. Replacing 5% of total energy intake from SFAs with isocaloric PUFAs was associated with decreased odds of hyperuricemia in men (0.69 (0.57, 0.84)) and in individuals (0.81 (0.71, 0.92)). Similar trends were observed in the substitution of SFAs with non-marine PUFAs in men (0.87 (0.80, 0.94)) and in all individuals (0.92 (0.88, 0.98)). Sensitivity analyses exhibited consistent results with primary analyses. CONCLUSION Higher dietary intake of n-3 PUFAs, n-6 PUFAs, LA, ALA, and non-marine PUFAs was associated with decreased hyperuricemia risk. These results support the recommendation to substitute SFAs with PUFAs in diet.
Collapse
Affiliation(s)
- Huimin Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guang Yang
- Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, Wuhan 430030, China
| | - Li Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, Wuhan 430030, China; State Environmental Protection Key Laboratory of Health Effects of Environmental Pollution, China; State Key Laboratory of Environment Health (Incubation), Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, Wuhan 430030, China
| | - Yanyan Li
- Shenzhen Center for Chronic Disease Control, 2021 Buxin Road, Shenzhen 518020, China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Environment & Health (Huazhong University of Science and Technology), Ministry of Education, Wuhan 430030, China; State Environmental Protection Key Laboratory of Health Effects of Environmental Pollution, China; State Key Laboratory of Environment Health (Incubation), Wuhan 430030, China; Hubei Key Laboratory of Food Nutrition and Safety, Wuhan 430030, China.
| | - Dongyan Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
14
|
Yang X, Liu D, Zhao X, Han Y, Zhang X, Zhou Q, Lv Q. Hyperuricemia drives intestinal barrier dysfunction by regulating gut microbiota. Heliyon 2024; 10:e36024. [PMID: 39224259 PMCID: PMC11367111 DOI: 10.1016/j.heliyon.2024.e36024] [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: 04/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Background Hyperuricemia elevates gut permeability; however, the risk of its influence on the compromised intestinal barrier is poorly understood. Aims This study was carried out, aiming to elucidate the orchestrators and disruptors of intestinal barrier in hyperuricemia. Methods A mouse model of hyperuricemia was induced by administering adenine and oteracil potassium to mice. Allopurinol was used to decrease uric acid level, and antibiotics were administered to mice to deplete gut microbiota. Intestinal permeability was assessed using FITC-labeled dextran. Changes in gut microbial community were analyzed through 16S rRNA sequencing. IL-1β and TNF-α levels were quantified using ELISA. The expression of tight junction protein genes, TLR4, p65 and IL-1β, was determined with Q-PCR and Western blotting. Results Allopurinol treatment effectively reduced intestinal permeability and serum TNF-α levels. Antibiotic treatment alleviated but not abolished intestinal permeability. Uric acid alone was insufficient to increase Coca2 monolayer permeability. Allopurinol treatment altered microbial composition and suppressed opportunistic infections. Re-establishing hyperuricemia in a germfree mouse model protected mice from intestinal injury. Allopurinol and antibiotic treatments reduced TLR4 and IL-1β expressions, increased occludin and claudin-1 expressions but suppressed NF-ĸB p65 signaling. However, removing gut microbiota aggravated lipid metabolic dysfunction. Conclusion Gut microbiota is a direct and specific cause for intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Xiaomin Yang
- Laboratory Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Dan Liu
- Laboratory Medicine, Qingdao Fuwai Cardiovascular Hospital, PR China
| | - Xiangzhong Zhao
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Yafei Han
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Xiao Zhang
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Quan Zhou
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| | - Qiulan Lv
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, 266003, PR China
| |
Collapse
|
15
|
Wang H, Zheng Y, Yang M, Wang L, Xu Y, You S, Mao N, Fan J, Ren S. Gut microecology: effective targets for natural products to modulate uric acid metabolism. Front Pharmacol 2024; 15:1446776. [PMID: 39263572 PMCID: PMC11387183 DOI: 10.3389/fphar.2024.1446776] [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: 06/10/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Gut microecology,the complex community consisting of microorganisms and their microenvironments in the gastrointestinal tract, plays a vital role in maintaining overall health and regulating various physiological and pathological processes. Recent studies have highlighted the significant impact of gut microecology on the regulation of uric acid metabolism. Natural products, including monomers, extracts, and traditional Chinese medicine formulations derived from natural sources such as plants, animals, and microorganisms, have also been investigated for their potential role in modulating uric acid metabolism. According to research, The stability of gut microecology is a crucial link for natural products to maintain healthy uric acid metabolism and reduce hyperuricemia-related diseases. Herein, we review the recent advanced evidence revealing the bidirectional regulation between gut microecology and uric acid metabolism. And separately summarize the key evidence of natural extracts and herbal formulations in regulating both aspects. In addition,we elucidated the important mechanisms of natural products in regulating uric acid metabolism and secondary diseases through gut microecology, especially by modulating the composition of gut microbiota, gut mucosal barrier, inflammatory response, purine catalyzation, and associated transporters. This review may offer a novel insight into uric acid and its associated disorders management and highlight a perspective for exploring its potential therapeutic drugs from natural products.
Collapse
Affiliation(s)
- Hui Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yixuan Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengfan Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yao Xu
- Chengdu Medical College, Chengdu, China
| | - Siqi You
- Chengdu Medical College, Chengdu, China
| | - Nan Mao
- Chengdu Medical College, Chengdu, China
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Junming Fan
- Chengdu Medical College, Chengdu, China
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Sichong Ren
- Chengdu Medical College, Chengdu, China
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- TCM Preventative Treatment Research Center of Chengdu Medical College, Chengdu, China
| |
Collapse
|
16
|
Yin X, Tong Q, Wang J, Wei J, Qin Z, Wu Y, Zhang R, Guan B, Qiu H. The impact of altered dietary adenine concentrations on the gut microbiota in Drosophila. Front Microbiol 2024; 15:1433155. [PMID: 39161604 PMCID: PMC11330887 DOI: 10.3389/fmicb.2024.1433155] [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: 05/15/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
The gut microbiota influences host metabolism and health, impacting diseases. Research into how diet affects gut microbiome dynamics in model organisms is crucial but underexplored. Herein, we examined how dietary adenine affects uric acid levels and the gut microbiota over five generations of Drosophila melanogaster. Wild-type W1118 flies consumed diets with various adenine concentrations (GC: 0%, GL: 0.05%, and GH: 0.10%), and their gut microbiota were assessed via Illumina MiSeq sequencing. Adenine intake significantly increased uric acid levels in the GH group > the GC group. Despite no significant differences in the alpha diversity indices, there were significant disparities in the gut microbiota health index (GMHI) and dysbiosis index (MDI) among the groups. Adenine concentrations significantly altered the diversity and composition of the gut microbiota. High adenine intake correlated with increased uric acid levels and microbial population shifts, notably affecting the abundances of Proteobacteria and Firmicutes. The gut microbiota phenotypes included mobile elements, gram-positive bacteria, biofilm-forming bacteria, and gram-negative bacteria. The significantly enriched KEGG pathways included ageing, carbohydrate metabolism, and the immune system. In conclusion, adenine intake increases uric acid levels, alters gut microbiota, and affects KEGG pathways in Drosophila across generations. This study highlights the impact of dietary adenine on uric acid levels and the gut microbiota, providing insights into intergenerational nutritional effects.
Collapse
Affiliation(s)
- Xianglin Yin
- School of Basic Medical Sciences, Jiamusi University, Heilongjiang, China
- School of Public Health, Jiamusi University, Heilongjiang, China
| | - Qing Tong
- School of Biology and Agriculture, Jiamusi University, Heilongjiang, China
| | - Jingtao Wang
- School of Basic Medical Sciences, Jiamusi University, Heilongjiang, China
| | - Jinfeng Wei
- School of Public Health, Jiamusi University, Heilongjiang, China
| | - Zhenbo Qin
- School of Public Health, Jiamusi University, Heilongjiang, China
| | - Yujie Wu
- School of Public Health, Jiamusi University, Heilongjiang, China
| | - Ruidi Zhang
- School of Public Health, Jiamusi University, Heilongjiang, China
| | - Baosheng Guan
- School of Public Health, Jiamusi University, Heilongjiang, China
| | - Hongbin Qiu
- School of Basic Medical Sciences, Jiamusi University, Heilongjiang, China
- School of Public Health, Jiamusi University, Heilongjiang, China
| |
Collapse
|
17
|
Jiang S, Xie S, Xie Z, Jiang W, Zhang H. Hyperuricemia Increases the Risk of Postoperative Recurrence in Chinese Patients with Chronic Rhinosinusitis. J Inflamm Res 2024; 17:2669-2679. [PMID: 38707953 PMCID: PMC11070159 DOI: 10.2147/jir.s457387] [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: 12/31/2023] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Background Elevated serum uric acid is crucial in the pathophysiology of chronic inflammatory diseases. However, its impact on chronic rhinosinusitis (CRS) recurrence risk is unknown. This study investigates the association between elevated serum uric acid and the risk of CRS recurrence. Methods A retrospective cohort study was conducted on 1004 CRS patients (including 638 males and 366 females) who received functional endoscopic sinus surgery. All patients were followed up for more than 2 years, and categorized into subgroups based on phenotype, gender, and postoperative recurrence. Cox regression analysis was performed to evaluate the associations between serum uric acid and the risk of CRS recurrence. Results After categorization, 104 males had hyperuricemia, and 54 females presented hyperuricemia. The rate of recurrent CRS in the hyperuricemia group was significantly higher compared to the non-hyperuricemia group in both males and females (P<0.05). In both male and female patients, the rate of hyperuricemia and uric acid levels were elevated in the recurrent CRS group in comparison with the non-recurrent CRS group (P<0.05). Unadjusted and adjusted Cox regression analysis demonstrated that serum uric acid was an independent risk factor for CRS recurrence (P<0.05). The receiver operator characteristic curve showed that serum uric acid was a potential biomarker for predicting the recurrence of CRS and its phenotypes in both genders (P<0.05). Conclusion There is a close relationship between elevated serum uric acid and the recurrence risk of CRS and its phenotypes, suggesting that serum uric acid may be a novel biomarker for predicting recurrent CRS.
Collapse
Affiliation(s)
- Sijie Jiang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Hunan Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Shaobing Xie
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Hunan Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Zhihai Xie
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Hunan Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Weihong Jiang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Hunan Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Hua Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Hunan Province Key Laboratory of Otolaryngology Critical Diseases, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Anatomy Laboratory of Division of Nose and Cranial Base, Clinical Anatomy Center of Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
18
|
Feng ZP, Wang XY, Xin HY, Huang SL, Huang HY, Xin Q, Zhang XH, Xin HW. Gut microbiota plays a significant role in gout. J Med Microbiol 2024; 73. [PMID: 38629677 DOI: 10.1099/jmm.0.001824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
With the development of social economy, the incidence of gout is increasing, which is closely related to people's increasingly rich diet. Eating a diet high in purine, fat, sugar and low-fibre for a long time further aggravates gout by affecting uric acid metabolism. The renal metabolism mechanism of uric acid has been thoroughly studied. To find a new treatment method for gout, increasing studies have recently been conducted on the mechanism of intestinal excretion, metabolism and absorption of uric acid. The most important research is the relationship between intestinal microbiota and the risk of gout. Gut microbiota represent bacteria that reside in a host's gastrointestinal tract. The composition of the gut microbiota is associated with protection against pathogen colonization and disease occurrence. This review focuses on how gut microbiota affects gout through uric acid and discusses the types of bacteria that may be involved in the occurrence and progression of gout. We also describe potential therapy for gout by restoring gut microbiota homeostasis and reducing uric acid levels. We hold the perspective that changing intestinal microbiota may become a vital method for effectively preventing or treating gout.
Collapse
Affiliation(s)
- Zhi-Peng Feng
- Key Laboratory of Research on Human Genetic Diseases Research at Universities of Inner Mongolia Autonomous Region, School of Basic Medicine, Chifeng University, Chifeng, Inner Mongolian Autonomous Region 024000, PR China
- Department of Gastroenterology, Yueyang Hospital Affiliated to Hunan Normal University, Yueyang, Hunan 414000, PR China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, PR China
| | - Xiao-Yan Wang
- The Doctoral Scientific Research Center, People's Hospital of Lianjiang, Guangdong 524400, PR China
- The Doctoral Scientific Research Center, People's Hospital of Lianjiang, Guangdong Medical University, Guangdong 524400, PR China
| | - Hong-Yi Xin
- The Doctoral Scientific Research Center, People's Hospital of Lianjiang, Guangdong 524400, PR China
- The Doctoral Scientific Research Center, People's Hospital of Lianjiang, Guangdong Medical University, Guangdong 524400, PR China
| | - Shao-Li Huang
- Clinical Laboratory, People's Hospital of Lianjiang, Guangdong 524400, PR China
| | - Hong-Yu Huang
- Department of Surgery, People's Hospital of Lianjiang, Guangdong 524400, PR China
| | - Qiang Xin
- Graduate School, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, PR China
- Department of Internal Medicine, Ulanqab General Hospital of Traditional Chinese Medicine and Mongolian Medicine, Hugeji Street South, Industry and Agriculture Street West, Jining New District, Ulanqab, Inner Mongolia 012000, PR China
| | - Xi-He Zhang
- The Doctoral Scientific Research Center, People's Hospital of Lianjiang, Guangdong 524400, PR China
- The Doctoral Scientific Research Center, People's Hospital of Lianjiang, Guangdong Medical University, Guangdong 524400, PR China
| | - Hong-Wu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, PR China
| |
Collapse
|
19
|
Qiu Y, Li C, Huang Y, Wu C, Li F, Zhang X, Xia D. Exploring the causal associations of micronutrients on urate levels and the risk of gout: A Mendelian randomization study. Clin Nutr 2024; 43:1001-1012. [PMID: 38484526 DOI: 10.1016/j.clnu.2024.03.003] [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: 08/08/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND & AIMS Growing evidence has indicated a potential association between micronutrient levels, urate levels, and the risk of gout. However, the causal association underlying these associations still remains uncertain. Previous observational studies and randomized controlled trials investigating the association between micronutrients, urate levels, and the risk of gout have been limited in their scope and depth. The aim of this study was to utilize Mendelian randomization (MR) to investigate the causal associations between genetically predicted micronutrient levels, urate levels, and the risk of gout. METHODS In this study, we conducted a comprehensive examination of 10 specific micronutrients (vitamin B6, vitamin B12, vitamin C, vitamin D, folate, calcium, iron, copper, zinc, and selenium) as potential exposures. Two-sample MR analyses were performed to explore their causal associations with urate levels and the risk of gout. In these analyses, gout data were collected from the Global Biobank Meta-Analysis Initiative (N = 1,069,839, N cases = 30,549) and urate levels data from CKDGen Consortium (N = 288,649) by utilizing publicly available summary statistics from independent cohorts of European ancestry. We performed inverse-variance weighted MR analyses as main analyses, along with a range of sensitivity analyses, such as MR-Egger, weighted median, simple mode, weighted mode, Steiger filtering, MR-PRESSO, and Radial MR analysis, to ensure the robustness of our findings. RESULTS The results of our study indicate that there were negative associations between serum vitamin B12 and urate levels, as well as serum folate and the risk of gout. Specifically, we found a negative association between vitamin B12 levels and urate levels, with a β coefficient of -0.324 (95% CI -0.0581 to -0.0066, P = 0.0137) per one standard deviation (SD) increase. Similarly, a negative association was observed between folate levels and gout risk, with an odds ratio of 0.8044 (95% CI 0.6637 to 0.9750, P = 0.0265) per one SD increase. On the other hand, we identified positive associations between serum calcium levels and both urate levels and the risk of gout. Specifically, there was a positive association between serum calcium levels and urate levels (β coefficient: 0.0994, 95% CI 0.0519 to 0.1468, P = 4.11E-05) per one SD increase. Furthermore, a positive association was found between serum calcium levels and the risk of gout, with an odds ratio of 1.1479 (95% CI 1.0460 to 1.2598, P = 0.0036) per one SD increase. These findings were robust in extensive sensitivity analyses. By employing MR-PRESSO and Radial MR to eliminate outliers, the observed associations have been reinforced. No clear associations were found between the other micronutrients and the urate levels, as well as the risk of gout. CONCLUSION Our findings provided evidence that there were negative associations between serum vitamin B12 and urate levels, as well as serum folate and the risk of gout, while positive associations existed between the serum calcium levels and urate levels, as well as the risk of gout.
Collapse
Affiliation(s)
- Yu Qiu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cantao Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxi Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fenfen Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoxi Zhang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Daozong Xia
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
20
|
Hussain A, Rui B, Ullah H, Dai P, Ahmad K, Yuan J, Liu Y, Li M. Limosilactobacillus reuteri HCS02-001 Attenuates Hyperuricemia through Gut Microbiota-Dependent Regulation of Uric Acid Biosynthesis and Excretion. Microorganisms 2024; 12:637. [PMID: 38674582 PMCID: PMC11052267 DOI: 10.3390/microorganisms12040637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Hyperuricemia is a prevalent metabolic disorder that arises from abnormal purine metabolism and reduced excretion of uric acid (UA). The gut microbiota plays a significant role in the biosynthesis and excretion of UA. Probiotics capable of purine degradation possess the potential to prevent hyperuricemia. Our study aimed to screen probiotics in areas with abundant dairy products and longevity populations in China, which could attenuate the level of UA and explore the underlying mechanism. In this study, twenty-three lactic acid bacteria isolated from healthy Chinese infant feces and traditional fermented foods such as hurood and lump milk were evaluated for the ability to tolerance acid, bile, artificial gastric juice, and artificial intestinal juice to determine the potential of the candidate strains as probiotics. Eight strains were identified as possessing superior tolerance to simulated intestinal conditions and were further analyzed by high-performance liquid chromatography (HPLC), revealing that Limosilactobacillus reuteri HCS02-001 (Lact-1) and Lacticaseibacillus paracasei HCS17-040 (Lact-2) possess the most potent ability to degrade purine nucleosides. The effect of Lact-1 and Lact-2 on hyperuricemia was evaluated by intervening with them in the potassium oxonate and adenine-induced hyperuricemia Balb/c mice model in vivo. Our results showed that the level of serum UA in hyperuricemic mice can be efficiently reduced via the oral administration of Lact-1 (p < 0.05). It significantly inhibited the levels of liver inflammatory cytokines and hepatic xanthine oxidase through a TLR4/MyD88/NF-κB pathway across the gut-liver axis. Furthermore, UA transporters ABCG2 and SLC2A9 were substantially upregulated by the intervention of this probiotic. Fecal ATP levels were significantly induced, while fecal xanthine dehydrogenase and allantoinase levels were increased following probiotics. RNA sequencing of HT-29 cells line treated with Lact-1 and its metabolites demonstrated significant regulation of pathways related to hyperuricemia. In summary, these findings demonstrate that Limosilactobacillus reuteri HCS02-001 possesses a capacity to ameliorate hyperuricemia by inhibiting UA biosynthesis via enhancing gastrointestinal barrier functions and promoting UA removal through the upregulation of urate transporters, thereby providing a basis for the probiotic formulation by targeting the gut microbiota.
Collapse
Affiliation(s)
- Akbar Hussain
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Binqi Rui
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Hayan Ullah
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Panpan Dai
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Kabir Ahmad
- Department of Physiology, Dalian Medical University, Dalian 116041, China;
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| |
Collapse
|
21
|
Qi X, Ma Y, Guan K, Zhao L, Ma Y, Wang R. Whey Protein Peptide Pro-Glu-Trp Ameliorates Hyperuricemia by Enhancing Intestinal Uric Acid Excretion, Modulating the Gut Microbiota, and Protecting the Intestinal Barrier in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2573-2584. [PMID: 38240209 DOI: 10.1021/acs.jafc.3c00984] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Hyperuricemia (HUA) is a metabolic disorder characterized by an increase in the concentrations of uric acid (UA) in the bloodstream, intricately linked to the onset and progression of numerous chronic diseases. The tripeptide Pro-Glu-Trp (PEW) was identified as a xanthine oxidase (XOD) inhibitory peptide derived from whey protein, which was previously shown to mitigate HUA by suppressing UA synthesis and enhancing renal UA excretion. However, the effects of PEW on the intestinal UA excretion pathway remain unclear. This study investigated the impact of PEW on alleviating HUA in rats from the perspective of intestinal UA transport, gut microbiota, and intestinal barrier. The results indicated that PEW inhibited the XOD activity in the serum, jejunum, and ileum, ameliorated intestinal morphology changes and oxidative stress, and upregulated the expression of ABCG2 and GLUT9 in the small intestine. PEW reversed gut microbiota dysbiosis by decreasing the abundance of harmful bacteria (e.g., Bacteroides, Alloprevotella, and Desulfovibrio) and increasing the abundance of beneficial microbes (e.g., Muribaculaceae, Lactobacillus, and Ruminococcus) and elevated the concentration of short-chain fatty acids. PEW upregulated the expression of occludin and ZO-1 and decreased serum IL-1β, IL-6, and TNF-α levels. Our findings suggested that PEW supplementation ameliorated HUA by enhancing intestinal UA excretion, modulating the gut microbiota, and restoring the intestinal barrier function.
Collapse
Affiliation(s)
- Xiaofen Qi
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education & Heilongjiang Provincial Key Laboratory of Ecological Restoration and Resource Utilization for Cold Region & Key Laboratory of Microbiology, College of Heilongjiang Province & School of Life Sciences, Heilongjiang University, Harbin 150080, China
| | - Yanfeng Ma
- Mengniu Hi-tech Dairy (Beijing) Co., Ltd., Beijing 101107, China
| | - Kaifang Guan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Le Zhao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Rongchun Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| |
Collapse
|
22
|
Lin X, Zou X, Hu B, Sheng D, Zhu T, Yin M, Xia H, Hu H, Liu H. Bi Xie Fen Qing Yin decoction alleviates potassium oxonate and adenine induced-hyperuricemic nephropathy in mice by modulating gut microbiota and intestinal metabolites. Biomed Pharmacother 2024; 170:116022. [PMID: 38147734 DOI: 10.1016/j.biopha.2023.116022] [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: 10/24/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 12/28/2023] Open
Abstract
This study aimed to evaluate the preventive effect of Bi Xie Fen Qing Yin (BXFQY) decoction on hyperuricemic nephropathy (HN). Using an HN mouse model induced by oral gavage of potassium oxonate and adenine, we found that BXFQY significantly reduced plasma uric acid levels and improved renal function. Further study shows that BXFQY suppressed the activation of the NLRP3 inflammasome and decreased the mRNA expressions of pro-inflammatory and fibrosis-associated factors in renal tissues of HN mice. Also, BXFQY prevented the damage to intestinal tissues of HN mice, indicative of suppressed colonic inflammation and increased gut barrier integrity. By 16 S rDNA sequencing, BXFQY significantly improved gut microbiota dysbiosis of HN mice. On the one hand, BXFQY down-regulated the abundance of some harmful bacteria, like Desulfovibrionaceae, Enterobacter, Helicobacter, and Desulfovibrio. On the other hand, BXFQY up-regulated the contents of several beneficial microbes, such as Ruminococcaceae, Clostridium sensu stricto 1, and Streptococcus. Using gas or liquid chromatography-mass spectrometry (GC/LC-MS) analysis, BXFQY reversed the changes in intestinal bacterial metabolites of HN mice, including indole and BAs. The depletion of intestinal flora from HN or HN plus BXFQY mice confirmed the significance of gut microbiota in BXFQY-initiated treatment of HN. In conclusion, BXFQY can alleviate renal inflammation and fibrosis of HN mice by modulating gut microbiota and intestinal metabolites. This study provides new insight into the underlying mechanism of BXFQY against HN.
Collapse
Affiliation(s)
- Xianghao Lin
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Xianning Medical College, Hubei University of Science and Technology, Xianning Avenue 88, Xianning 437100, PR China
| | - Xiaojuan Zou
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Dongyun Sheng
- Department of Traditional Chinese Medicine, General Hospital of China Resources WISCO, Metallurgy Avenue 29, Wuhan 430080, PR China
| | - Tianxiang Zhu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Mingzhu Yin
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Hui Xia
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Haiming Hu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| |
Collapse
|
23
|
Lv Q, Zhou J, Wang C, Yang X, Han Y, Zhou Q, Yao R, Sui A. A dynamics association study of gut barrier and microbiota in hyperuricemia. Front Microbiol 2023; 14:1287468. [PMID: 38088975 PMCID: PMC10711221 DOI: 10.3389/fmicb.2023.1287468] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/09/2023] [Indexed: 02/28/2024] Open
Abstract
Introduction The intricate interplay between gut microbiota and hyperuricemia remains a subject of growing interest. However, existing studies only provided snapshots of the gut microbiome at single time points, the temporal dynamics of gut microbiota alterations during hyperuricemia progression and the intricate interplay between the gut barrier and microbiota remain underexplored. Our investigation revealed compelling insights into the dynamic changes in both gut microbiota and intestinal barrier function throughout the course of hyperuricemia. Methods The hyperuricemia mice (HY) were given intragastric administration of adenine and potassium oxalate. Gut microbiota was analyzed by 16S rRNA sequencing at 3, 7, 14, and 21 days after the start of the modeling process. Intestinal permeability as well as LPS, TNF-α, and IL-1β levels were measured at 3, 7, 14, and 21 days. Results We discovered that shifts in microbial community composition occur prior to the onset of hyperuricemia, key bacterial Bacteroidaceae, Bacteroides, and Blautia exhibited reduced levels, potentially fueling microbial dysbiosis as the disease progresses. During the course of hyperuricemia, the dynamic fluctuations in both uric acid levels and intestinal barrier function was accompanied with the depletion of key beneficial bacteria, including Prevotellaceae, Muribaculum, Parabacteroides, Akkermansia, and Bacteroides, and coincided with an increase in pathogenic bacteria such as Oscillibacter and Ruminiclostridium. This microbial community shift likely contributed to elevated lipopolysaccharide (LPS) and pro-inflammatory cytokine levels, ultimately promoting metabolic inflammation. The decline of Burkholderiaceae and Parasutterella was inversely related to uric acid levels, Conversely, key families Ruminococcaceae, Family_XIII, genera Anaeroplasma exhibited positive correlations with uric acid levels. Akkermansiaceae and Bacteroidaceae demonstrating negative correlations, while LPS-containing microbiota such as Desulfovibrio and Enterorhabdus exhibited positive correlations with intestinal permeability. Conclusion In summary, this study offers a dynamic perspective on the complex interplay between gut microbiota, uric acid levels, and intestinal barrier function during hyperuricemia progression. Our study suggested that Ruminiclostridium, Bacteroides, Akkermansiaceae, Bilophila, Burkholderiaceae and Parasutterella were the key bacteria that play vital rols in the progress of hyperuricemia and compromised intestinal barrier, which provide a potential avenue for therapeutic interventions in hyperuricemia.
Collapse
Affiliation(s)
- Qiulan Lv
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun Zhou
- Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changyao Wang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaomin Yang
- Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yafei Han
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Quan Zhou
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruyong Yao
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aihua Sui
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Li JG, Gao LL, Wang CC, Tu JM, Chen WH, Wu XL, Wu JX. Colonization of multidrug-resistant Gram-negative bacteria increases risk of surgical site infection after hemorrhoidectomy: a cross-sectional study of two centers in southern China. Int J Colorectal Dis 2023; 38:243. [PMID: 37779168 PMCID: PMC10543959 DOI: 10.1007/s00384-023-04535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
PURPOSE The present study aims to determine the rectoanal colonization rate and risk factors for the colonization of present multidrug-resistant bacteria (MDRBs). In addition, the relationship between MDRB colonization and surgical site infection (SSI) following hemorrhoidectomy was explored. METHODS A cross-sectional study was conducted in the Department of Colorectal Surgery of two hospitals. Patients with hemorrhoid disease, who underwent hemorrhoidectomy, were included. The pre-surgical screening of multidrug-resistant Gram-negative bacteria (MDR-GNB) colonization was performed using rectal swabs on the day of admission. Then, the MDRB colonization rate was determined through the rectal swab. Logistic regression models were established to determine the risk factors for MDRB colonization and SSI after hemorrhoidectomy. A p-value of < 0.05 was considered statistically significant. RESULTS A total of 432 patients met the inclusion criteria, and the MDRB colonization prevalence was 21.06% (91/432). The independent risk factors for MDRB colonization were as follows: patients who received ≥ 2 categories of antibiotic treatment within 3 months (odds ratio (OR): 3.714, 95% confidence interval (CI): 1.436-9.605, p = 0.007), patients with inflammatory bowel disease (IBD; OR: 6.746, 95% CI: 2.361-19.608, p < 0.001), and patients with high serum uric acid (OR: 1.006, 95% CI: 1.001-1.010, p = 0.017). Furthermore, 41.57% (37/89) of MDRB carriers and 1.81% (6/332) of non-carriers developed SSIs, with a total incidence of 10.21% (43/421). Based on the multivariable model, the rectoanal colonization of MDRBs (OR: 32.087, 95% CI: 12.052-85.424, p < 0.001) and hemoglobin < 100 g/L (OR: 4.130, 95% CI: 1.556-10.960, p = 0.004) were independently associated with SSI after hemorrhoidectomy. CONCLUSION The rectoanal colonization rate of MDRBs in hemorrhoid patients is high, and this was identified as an independent risk factor for SSI after hemorrhoidectomy.
Collapse
Affiliation(s)
- Jian-Guo Li
- Department of Colorectal Surgery, First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Li-Lian Gao
- Department of Colorectal Surgery, First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Cun-Chuan Wang
- Department of Colorectal Surgery, First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Jia-Min Tu
- Department of Colorectal Surgery, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen, China
| | - Wen-Hui Chen
- Department of Colorectal Surgery, First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Xiang-Lin Wu
- Department of Colorectal Surgery, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen, China
| | - Jin-Xia Wu
- Department of Colorectal Surgery, University of Chinese Academy of Sciences Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
25
|
Dang K, Zhang N, Gao H, Wang G, Liang H, Xue M. Influence of intestinal microecology in the development of gout or hyperuricemia and the potential therapeutic targets. Int J Rheum Dis 2023; 26:1911-1922. [PMID: 37606177 DOI: 10.1111/1756-185x.14888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
Gout and hyperuricemia are common metabolic diseases. Patients with purine metabolism disorder and/or decreased uric acid excretion showed increased uric acid levels in the blood. The increase of uric acid in the blood leads to the deposition of urate crystals in tissues, joints, and kidneys, and causes gout. Recent studies have revealed that imbalance of the intestinal microecology is closely related to the occurrence and development of hyperuricemia and gout. Disorder of the intestinal flora often occurs in patients with gout, and high purine and high fructose may induce the disorder of intestinal flora. Short-chain fatty acids and endotoxins produced by intestinal bacteria are closely related to the inflammatory response of gout. This article summarizes the characteristics of intestinal microecology in patients or animal models with hyperuricemia or gout, and explores the relationship between intestinal microecology and gout or hyperuricemia from the aspect of the intestinal barrier, intestinal microorganisms, intestinal metabolites, and intestinal immune system. We also review the current status of hyperuricemia treatment by targeting intestinal microecology.
Collapse
Affiliation(s)
- Kai Dang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Haiqi Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Guifa Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Hui Liang
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Hamid O, Alsabbagh Alchirazi K, Eltelbany A, Nanah R, Regueiro M. Increased prevalence of gout in patients with inflammatory bowel disease: A population-based study. JGH Open 2023; 7:640-644. [PMID: 37744707 PMCID: PMC10517438 DOI: 10.1002/jgh3.12963] [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: 03/04/2023] [Revised: 05/14/2023] [Accepted: 08/19/2023] [Indexed: 09/26/2023]
Abstract
Background and Aim Arthritis is a recognized extra-intestinal manifestation of inflammatory bowel disease (IBD). Studies show altered uric acid metabolism in IBD. This study aims to investigate the association between IBD and gout. Methods We used a multi-center database (Explorys Inc.) consisting of data from several US healthcare systems. We identified adults diagnosed with Crohn's disease (CD) and ulcerative colitis (UC) between 1999 and 2022. In this cohort, we identified patients diagnosed with gout. We collected demographic data and identified patients diagnosed with IBD-associated arthritis and those who had intestinal resection. Risk factors associated with gout were collected. Multivariate analysis was used. Results Out of the 69 260 780 patients in the database, we identified 209 020 patients with UC (0.30%) of whom 9130 had gout (4.3%). Additionally, 249 480 had CD (0.36%) of whom 14 000 had gout (5.61%). Males were more prevalent in the UC and gout group than in the CD and gout group (58% vs 51%). After adjustment, CD was significantly associated with gout (odds ratio [OR] 1.68, confidence interval [CI]: 1.60-1.75). UC was also significantly associated with gout (OR 1.38, CI: 1.31-1.44). In subgroup analysis with intestinal resection, CD patients who had intestinal resection had higher association with gout versus those without surgery (OR 2.34, CI: 2.25-2.43). Similar increase was observed in the UC group with intestinal resection (OR 1.53, CI: 1.49-1.56). Conclusion IBD is strongly associated with gout, with higher correlation observed with CD. Intestinal resection is associated with an increase in the risk of gout. Patients with IBD who present with new-onset arthritis should be investigated for gout.
Collapse
Affiliation(s)
- Osama Hamid
- Department of Hospital MedicineCleveland ClinicClevelandOhioUSA
| | | | - Ahmed Eltelbany
- Department of Hospital MedicineCleveland ClinicClevelandOhioUSA
| | - Rama Nanah
- Department of Hospital MedicineCleveland ClinicClevelandOhioUSA
| | - Miguel Regueiro
- Department of Gastroenterology and HepatologyCleveland ClinicClevelandOhioUSA
- Department of MedicineCleveland Clinic Lerner College of Medicine, Case Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
27
|
Zhang Y, Wang S, Dai X, Liu T, Liu Y, Shi H, Yin J, Xu T, Zhang Y, Zhao D, Sukhorukov V, Orekhov AN, Gao S, Wang L, Zhang D. Simiao San alleviates hyperuricemia and kidney inflammation by inhibiting NLRP3 inflammasome and JAK2/STAT3 signaling in hyperuricemia mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116530. [PMID: 37098372 DOI: 10.1016/j.jep.2023.116530] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Simiao San (SmS), a famous traditional Chinese formula, is clinically used to treat patients with hyperuricemia (HUA). However, its mechanism of action on lowering uric acid (UA) and inhibiting inflammation still deserves further investigation. AIM OF THE STUDY To examine the effect and its possible underlying mechanism of SmS on UA metabolism and kidney injury in HUA mouse. MATERIALS AND METHODS The HUA mouse model was constructed with the combined administration of both potassium oxalate and hypoxanthine. The effects of SmS on UA, xanthine oxidase (XOD), creatinine (CRE), blood urea nitrogen (BUN), interleukin-10 (IL-10), interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) were determined by ELISA or biochemical assays. Hematoxylin and eosin (H&E) was used to observe pathological alterations in the kidneys of HUA mice. The expression levels of organic anion transporter 1 (OAT1), recombinant urate transporter 1 (URAT1), glucose transporter 9 (GLUT9), nucleotide binding domain and leucine rich repeat pyrin domain containing 3 (NLRP3), Cleaved-Caspase 1, apoptosis-associated speck like protein (ASC), nuclear factor kappa-B (NF-κB), IL-6, janus kinase 2 (JAK2), phosphor (P)-JAK2, signal transducers and activators of transcription 3 (STAT3), P-STAT3, suppressor of cytokine signaling 3 (SOCS3) were examined by Western blot and/or immunohistochemical (IHC) staining. The major ingredients in SmS were identified by a HPLC-MS assay. RESULTS HUA mouse exhibited an elevation in serum levels of UA, BUN, CRE, XOD, and the ratio of urinary albumin to creatinine (UACR), and a decline in urine levels of UA and CRE. In addition, HUA induces pro-inflammatory microenvironment in mouse, including an increase in serum levels of IL-1β, IL-6, and TNF-α, and renal expressions of URAT1, GULT9, NLRP3, ASC, Cleaved-Caspase1, P-JAK2/JAK2, P-STAT3/STAT3, and SOCS3, and a decrease in serum IL-10 level and renal OAT1 expression as well as a disorganization of kidney pathological microstructure. In contrast, SmS intervention reversed these alterations in HUA mouse. CONCLUSION SmS could alleviate hyperuricemia and renal inflammation in HUA mouse. The action mechanisms behind these alterations may be associated with a limitation of the NLRP3 inflammasome and JAK2/STAT3 signaling pathways.
Collapse
Affiliation(s)
- Yueyi Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shan Wang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xuan Dai
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Tianyuan Liu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yage Liu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Hanfen Shi
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jiyuan Yin
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Tianshu Xu
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yanfei Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Dandan Zhao
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Vasily Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 125315, Russia.
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, 125315, Russia.
| | - Sihua Gao
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Lili Wang
- Department of TCM Pharmacology, Chinese Material Medica School, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Dongwei Zhang
- Diabetes Research Center, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
28
|
Shirvani-Rad S, Khatibzade-Nasari N, Ejtahed HS, Larijani B. Exploring the role of gut microbiota dysbiosis in gout pathogenesis: a systematic review. Front Med (Lausanne) 2023; 10:1163778. [PMID: 37265486 PMCID: PMC10230090 DOI: 10.3389/fmed.2023.1163778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Objectives Gut dysbiosis is believed to be one of the several mechanisms that are involved in the pathogenesis of gout. This systematic review aimed to summarize the role of gut dysbiosis in gout disease and uncover the underlying mechanisms. Methods A comprehensive search was conducted on PubMed, Web of Science, and Scopus databases up to October 2021. Animal studies and human observational studies, including case-control, cross-sectional, and cohort studies assessing the association between gut microbiota composition and gout were included. The quality of included studies has been evaluated using the Newcastle-Ottawa Quality Assessment scale (NOS) and the SYRCLE's risk of bias tool. Results Initially, we found 274 studies among which 15 studies were included in this systematic review. Of them, 10 studies were conducted on humans and 5 studies were conducted on animals. Increased abundance of Alistipes and decreased abundance of Enterobacteriaceae alters purine metabolism, thereby aggravating gout condition. Moreover, a higher abundance of Phascolarctobacterium and Bacteroides in gout modulates enzymatic activity in purine metabolism. Butyrate-producing bacteria such as Faecalibacterium, prausnitzii, Oscillibacter, Butyricicoccus, and Bifidobacterium have higher abundance in healthy controls compared to gout patients, suggesting the anti-inflammatory and anti-microbial role of short-chain fatty acids (SCFAs). Lipopolysaccharides (LPS)-releasing bacteria, such as Enterobacteriaceae, Prevotella, and Bacteroides, are also involved in the pathogenesis of gout disease by stimulating the innate immune system. Conclusion Exploring the role of gut dysbiosis in gout and the underlying mechanisms can help develop microbiota-modulating therapies for gout.
Collapse
Affiliation(s)
- Salman Shirvani-Rad
- Microbiota Research Group, Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Qeshm International Medical Sciences Branch, Islamic Azad University, Qeshm, Iran
| | - Niloufar Khatibzade-Nasari
- Faculty of Medicine, Qeshm International Medical Sciences Branch, Islamic Azad University, Qeshm, Iran
- Young Researchers and Elite Club, Qeshm International Medical Sciences Branch, Islamic Azad University, Qeshm, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Brunetti L, Wang L, Wassef A, Gong Y, Brinker A, Buckley B, Lipsky PE, Ondar P, Poiani G, Zhao L, Kong AN, Schlesinger N. Pharmacokinetics and Pharmacodynamics of Anthocyanins after Administration of Tart Cherry Juice to Individuals with Gout. Mol Nutr Food Res 2023; 67:e2200550. [PMID: 36843307 DOI: 10.1002/mnfr.202200550] [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: 08/18/2022] [Revised: 12/21/2022] [Indexed: 02/28/2023]
Abstract
SCOPE Tart cherries (TCs) contain high levels of anthocyanins that exert potent antioxidant and antiinflammatory effects and potentially benefit individuals with gout. METHODS AND RESULTS This study aims to quantitate the major anthocyanins in TC Juice Concentrate (TCJC) and identify the pharmacokinetic (PK) and pharmacodynamic (PD) parameters of the major anthocyanin cyanidin-3-glucosylrutinoside (C3GR). A PK-PD study enrolling human subjects with a history of gout is performed. Subjects are randomized to receive either 60 or 120 mL of TCJC. Anthocyanins are quantitated using liquid chromatography-mass spectroscopy (LCMS). Antioxidant and antiinflammatory mRNA expression is measured using real-time qPCR before and after the administration of TCJC. A population PK model (popPK) is fit to the experimental data, and an indirect PD model (IDR) is constructed in Monolix. CONCLUSION Of the bioavailable anthocyanins, C3GR achieves the highest plasma concentration in a dose-dependent manner. A popPK predicts anthocyanin exposure, and an IDR produces reasonable approximations of PD effects.
Collapse
Affiliation(s)
- Luigi Brunetti
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, 08854, USA
- Robert Wood Johnson University Hospital Somerset, Somerville, NJ, 08876, USA
| | - Lujing Wang
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, 08854, USA
| | - Andrew Wassef
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, 08854, USA
| | - Yongjia Gong
- New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ, 08901, USA
| | - Anita Brinker
- Environmental and Occupational Health Sciences Institute (EOSHI), Rutgers University, Piscataway, NJ, 08854, USA
| | - Brian Buckley
- Environmental and Occupational Health Sciences Institute (EOSHI), Rutgers University, Piscataway, NJ, 08854, USA
| | - Peter E Lipsky
- RILITE Research Institute and AMPEL BioSolutions, Charlottesville, VA, 22902, USA
| | - Patricia Ondar
- Robert Wood Johnson University Hospital, New Brunswick, NJ, 08901, USA
| | - George Poiani
- Robert Wood Johnson University Hospital Somerset, Somerville, NJ, 08876, USA
| | - Liping Zhao
- New Jersey Institute for Food, Nutrition, and Health, New Brunswick, NJ, 08901, USA
| | - Ah-Ng Kong
- Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, 08854, USA
| | - Naomi Schlesinger
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| |
Collapse
|
30
|
Hou T, Dai H, Wang Q, Hou Y, Zhang X, Lin H, Wang S, Li M, Zhao Z, Lu J, Xu Y, Chen Y, Gu Y, Zheng J, Wang T, Wang W, Bi Y, Ning G, Xu M. Dissecting the causal effect between gut microbiota, DHA, and urate metabolism: A large-scale bidirectional Mendelian randomization. Front Immunol 2023; 14:1148591. [PMID: 37063923 PMCID: PMC10097983 DOI: 10.3389/fimmu.2023.1148591] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023] Open
Abstract
ObjectivesOur aim was to investigate the interactive causal effects between gut microbiota and host urate metabolism and explore the underlying mechanism using genetic methods.MethodsWe extracted summary statistics from the abundance of 211 microbiota taxa from the MiBioGen (N =18,340), 205 microbiota metabolism pathways from the Dutch Microbiome Project (N =7738), gout from the Global Biobank Meta-analysis Initiative (N =1,448,128), urate from CKDGen (N =288,649), and replication datasets from the Global Urate Genetics Consortium (N gout =69,374; N urate =110,347). We used linkage disequilibrium score regression and bidirectional Mendelian randomization (MR) to detect genetic causality between microbiota and gout/urate. Mediation MR and colocalization were performed to investigate potential mediators in the association between microbiota and urate metabolism.ResultsTwo taxa had a common causal effect on both gout and urate, whereas the Victivallaceae family was replicable. Six taxa were commonly affected by both gout and urate, whereas the Ruminococcus gnavus group genus was replicable. Genetic correlation supported significant results in MR. Two microbiota metabolic pathways were commonly affected by gout and urate. Mediation analysis indicated that the Bifidobacteriales order and Bifidobacteriaceae family had protective effects on urate mediated by increasing docosahexaenoic acid. These two bacteria shared a common causal variant rs182549 with both docosahexaenoic acid and urate, which was located within MCM6/LCT locus.ConclusionsGut microbiota and host urate metabolism had a bidirectional causal association, implicating the critical role of host-microbiota crosstalk in hyperuricemic patients. Changes in gut microbiota can not only ameliorate host urate metabolism but also become a foreboding indicator of urate metabolic diseases.
Collapse
Affiliation(s)
- Tianzhichao Hou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huajie Dai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanan Hou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyun Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyun Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Min Xu,
| |
Collapse
|
31
|
He L, Tang W, Huang L, Zhou W, Huang S, Zou L, Yuan L, Men D, Chen S, Hu Y. Rational design of a genome-based insulated system in Escherichia coli facilitates heterologous uricase expression for hyperuricemia treatment. Bioeng Transl Med 2023; 8:e10449. [PMID: 36925686 PMCID: PMC10013758 DOI: 10.1002/btm2.10449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/18/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Hyperuricemia is a prevalent disease worldwide that is characterized by elevated urate levels in the blood owing to purine metabolic disorders, which can result in gout and comorbidities. To facilitate the treatment of hyperuricemia through the uricolysis, we engineered a probiotic Escherichia coli Nissle 1917 (EcN) named EcN C6 by inserting an FtsP-uricase cassette into an "insulated site" located between the uspG and ahpF genes. Expression of FtsP-uricase in this insulated region did not influence the probiotic properties or global gene transcription of EcN but strongly increased the enzymatic activity for urate degeneration, suggesting that the genome-based insulated system is an ideal strategy for EcN modification. Oral administration of EcN C6 successfully alleviated hyperuricemia, related symptoms and gut microbiota in a purine-rich food-induced hyperuricemia rat model and a uox-knockout mouse model. Together, our study provides an insulated site for heterologous gene expression in EcN strain and a recombinant EcN C6 strain as a safe and effective therapeutic candidate for hyperuricemia treatment.
Collapse
Affiliation(s)
- Lina He
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Wei Tang
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
| | - Ling Huang
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Wei Zhou
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shaojia Huang
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Linxuan Zou
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Lisha Yuan
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Dong Men
- State Key Laboratory of VirologyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
| | - Shiyun Chen
- CAS Key Laboratory of Special Pathogens and BiosafetyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
| | - Yangbo Hu
- State Key Laboratory of VirologyWuhan Institute of Virology, Chinese Academy of SciencesWuhanChina
- Hubei Jiangxia LaboratoryWuhanChina
| |
Collapse
|
32
|
Wang Z, Song L, Li X, Xiao Y, Huang Y, Zhang Y, Li J, Li M, Ren Z. Lactiplantibacillus pentosus P2020 protects the hyperuricemia and renal inflammation in mice. Front Nutr 2023; 10:1094483. [PMID: 36891165 PMCID: PMC9987516 DOI: 10.3389/fnut.2023.1094483] [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: 11/10/2022] [Accepted: 01/10/2023] [Indexed: 02/22/2023] Open
Abstract
Introduction Hyperuricemia (HUA) is a common metabolic disease, and its prevalence has been increasing worldwide. Pharmaceutical drugs have been used for controlling HUA but they all have certain side effects, which thus calls for discovering alternative options including using treatment of probiotics to prevent the development of HUA. Methods We established HUA mice model induced by potassium oxonate and adenine and performed in vivo experiments to verify the ability to lower serum uric acid of Lactiplantibacillus pentosus P2020 (LPP), a probiotics stain extracted from Chinese pickle. We also tried to discussed the underlying mechanisms. Results Oral administration with LPP significantly decreased serum uric acid and reduced renal inflammatory response by downregulating multiple inflammation pathways including NK-kB, MAPK, and TNFα. We also found that LPP administration significantly promoted uric acid excretion by regulating expression of transporters in the kidney and ileum. In addition, LPP intake improved intestinal barrier function and modulated the composition of gut microbiota. Discussion These results suggest that probiotics LPP may have a promising potential to protect against development of HUA and HUA-related renal damage, and its working mechanisms involve regulation of inflammation pathways and expression of transporters in the kidney and ileum.
Collapse
Affiliation(s)
- Zhihuan Wang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Liqiong Song
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Xianping Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuchun Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanming Huang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yue Zhang
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jintong Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingding Li
- Maiyata Institute for Beneficial Bacteria, Shaoxing, Zhejiang, China
| | - Zhihong Ren
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Huang Z, Zhang W, An Q, Lang Y, Liu Y, Fan H, Chen H. Exploration of the anti-hyperuricemia effect of TongFengTangSan (TFTS) by UPLC-Q-TOF/MS-based non-targeted metabonomics. Chin Med 2023; 18:17. [PMID: 36797795 PMCID: PMC9933412 DOI: 10.1186/s13020-023-00716-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 01/28/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND TongFengTangSan (TFTS) is a commonly used Tibetan prescription for gout treatment. Previously, TFTS (CF) was confirmed to have a significant uric acid-lowering effect. However, the anti-hyperuricemia mechanisms and the main active fractions remain unclear. The current study aimed to investigate the anti-hyperuricemia mechanism using metabolomics and confirm the active CF fraction. METHODS The hyperuricemia model was established through intraperitoneal injection containing 100 mg/kg potassium oxonate and 150 mg/kg hypoxanthine by gavage. We used serum uric acid (sUA), creatinine (CRE), blood urea nitrogen (BUN), xanthine oxidase (XOD) activity, interleukin-6 (IL-6) and interleukin-1β (IL-1β) as indicators to evaluate the efficacy of CF and the four fractions (SX, CF30, CF60, and CF90). The anti-hyperuricemia mechanism of CF was considered through non-targeted metabolomics depending on the UPLC-Q-TOF-MS technology. Principle component analysis (PCA) and orthogonal partial least squares-discriminant analysis (OPLS-DA) helped explore the potential biomarkers in hyperuricemia. Moreover, the differential metabolites and metabolic pathways regulated by CF and four fractions were also assessed. RESULTS CF revealed a significant anti-hyperuricemia effect by down-regulating the level of sUA, sCRE, sIL-1β, and XOD. SX, CF30, CF60, and CF90 differed in the anti-hyperuricemia effect. Only CF60 significantly lowered the sUA level among the four fractions, and it could be the main efficacy fraction of TFTS. Forty-three differential metabolites were identified in hyperuricemia rats from plasma and kidney. Pathway analysis demonstrated that seven pathways were disrupted among hyperuricemia rats. CF reversed 19 metabolites in hyperuricemia rats and exerted an anti-hyperuricemia effect by regulating purine metabolism. CF60 was the main active fraction of TFTS and exerted a similar effect of CF by regulating purine metabolism. CONCLUSIONS CF and CF60 could exert an anti-hyperuricemia effect by regulating the abnormal purine metabolism because of hyperuricemia while improving intestinal and renal function. CF60 could be the main active fraction of TFTS.
Collapse
Affiliation(s)
- Zhichao Huang
- grid.411868.20000 0004 1798 0690Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330004 China
| | - Wugang Zhang
- grid.411868.20000 0004 1798 0690Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330004 China ,grid.411868.20000 0004 1798 0690State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006 China
| | - Qiong An
- grid.411868.20000 0004 1798 0690Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330004 China
| | - Yifan Lang
- grid.411868.20000 0004 1798 0690State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006 China
| | - Ye Liu
- grid.411868.20000 0004 1798 0690State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006 China
| | - Huifang Fan
- grid.411868.20000 0004 1798 0690State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006 China
| | - Haifang Chen
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, China.
| |
Collapse
|
34
|
Global status and trends in gout research from 2012 to 2021: a bibliometric and visual analysis. Clin Rheumatol 2023; 42:1371-1388. [PMID: 36662336 PMCID: PMC9852810 DOI: 10.1007/s10067-023-06508-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/28/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND Gout is the most common inflammatory arthritis with an increasing prevalence and incidence across the globe. We aimed to provide a comprehensive and systematic knowledge map of gout research to determine its current status and trends over the past decade. METHODS Publications on gout research were obtained from the Web of Science Core Collection (WOSCC) database. Bibliometric R, VOSviewer, and Citespace were employed to analyze the eligible literature. RESULTS A total of 5535 publications concerning gout research between 2012 and 2021 were included. Most publications and citations both numerically came from China. The strongest international cooperation belonged to the USA. The University of Auckland was the most productive institution with a leading place in research collaboration. The prime funding agency was the National Natural Science Foundation of China. Most papers were published in Clinical Rheumatology. Annals of the Rheumatic Diseases achieved the highest number of citations, H-index and IF, which showed the most excellent comprehensive strength. The individual author with the most paper authorship was Dalbeth Nicola with 241 publications and 46 H-index. Keywords and co-citation analysis discovered that pathological mechanism remains the future hotspot in gout research. It may involve gout connection with gut microbiota, NLRP3 inflammasome, xanthine oxidase, and urate-transporter ABCG2. In addition, besides metabolic diseases, the relationship between gout and heart failure may need more attention. CONCLUSION This study clarified the current status and research frontier in gout over the past decade, which would provide valuable research references for later researchers. Key Points •We disclosed the current status and frontier directions of gout over the past 10 years worldwide. •We identified future hotspots of gout research, including gout connection with gut microbiota, NLRP3 inflammasome, xanthine oxidase, and urate-transporter ABCG2. •We discovered that the relationship between gout and heart status would be the research frontier.
Collapse
|
35
|
Xie D, Shen Y, Su E, Du L, Xie J, Wei D. Anti-Hyperuricemic, Nephroprotective, and Gut Microbiota Regulative Effects of Separated Hydrolysate of α-Lactalbumin on Potassium Oxonate- and Hypoxanthine-Induced Hyperuricemic Mice. Mol Nutr Food Res 2023; 67:e2200162. [PMID: 36308034 DOI: 10.1002/mnfr.202200162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 09/29/2022] [Indexed: 01/19/2023]
Abstract
SCOPE This study aims to investigate the anti-hyperuricemic and nephroprotective effects and the potential mechanisms of the separated gastrointestinal hydrolysates of α-lactalbumin on hyperuricemic mice. METHODS AND RESULTS The gastrointestinal hydrolysate of α-lactalbumin, the hydrolysate fraction with molecular weight (MW) < 3 kDa (LH-3k), and the fragments with smallest MW among LH-3K harvested through dextran gel chromatography (F5) are used. Hyperuricemia mice are induced via daily oral gavage of potassium oxonate and hypoxanthine. F5 displays the highest in vitro xanthine oxidase (XO) inhibition among all the fractions separated from LH-3k. Oral administration of F5 significantly reduces the levels of serum uric acid (UA), creatinine, and urea nitrogen. F5 treatment could ameliorate kidney injury through alleviating oxidative stress and inflammation. F5 alleviates hyperuricemia in mice by inhibiting hepatic XO activity and regulating the expression of renal urate transporters. Gut microbiota analysis illustrates that F5 administration increases the abundance of some SCFAs producers, and inhibits the growth of hyperuricemia and inflammation associated genera. LH-3k exhibits similar effects but does not show significance as those of the F5 fraction. CONCLUSION The anti-hyperuricemia and nephroprotective functions of F5 are mediated by inhibiting hepatic XO activity, ameliorating oxidative stress and inflammation, regulating renal urate transporters, and modulating the gut microbiota in hyperuricemic mice.
Collapse
Affiliation(s)
- Dewei Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yaling Shen
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Erzheng Su
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, 210037, P. R. China
| | - Lei Du
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Jingli Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, P. R. China.,Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, P. R. China
| | - Dongzhi Wei
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, P. R. China.,Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, P. R. China
| |
Collapse
|
36
|
Kawakami Y, Mazuka M, Yasuda A, Sato M, Hosaka T, Arai H. Acute effect of fructose, sucrose, and isomaltulose on uric acid metabolism in healthy participants. J Clin Biochem Nutr 2023; 72:61-67. [PMID: 36777082 PMCID: PMC9899922 DOI: 10.3164/jcbn.22-41] [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: 04/07/2022] [Accepted: 07/02/2022] [Indexed: 01/01/2023] Open
Abstract
Fructose is associated with hyperuricemia and gout development. Focusing on fructose and fructose-containing disaccharides, we investigated the effects of three different types of carbohydrates (fructose, sucrose, and isomaltulose) on uric acid metabolism and gene expression profiling in peripheral white blood cells. In a randomized crossover study, ten healthy participants ingested test drinks of fructose, sucrose, and isomaltulose, each containing 25 g of fructose. Plasma glucose, serum and urine uric acid, and xanthine/hypoxanthine concentrations were measured. Microarray analysis in peripheral white blood cells and real-time reverse transcription polymerase chain reaction were examined at 0 and 120 in after the intake of test drinks. Serum uric acid concentrations for group fructose were significantly higher than group sucrose at 30-120 min and were significantly higher than those for group isomaltulose at 30-240 min. Several genes involved in the "nuclear factor-kappa B signaling pathway" were markedly changed in group fructose. No significant differences in the mRNA expression levels of tumor necrosis factor, nuclear factor-kappa B, interleukin-1β, and interleukin-18 were noted. This study indicated that fructose intake (monosaccharide) elevated serum uric acid concentrations compared with disaccharide intake. Differences in the quality of carbohydrates might reduce the rapid increase of postprandial serum uric acid concentrations.
Collapse
Affiliation(s)
- Yuka Kawakami
- Laboratory of Clinical Nutrition and Management, Graduate Division of Nutritional and Environmental Sciences, and Graduate School of Integrated Pharmaceutical and Nutritional Sciences, The University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Megumi Mazuka
- Laboratory of Clinical Nutrition and Management, Graduate Division of Nutritional and Environmental Sciences, and Graduate School of Integrated Pharmaceutical and Nutritional Sciences, The University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Arisa Yasuda
- Laboratory of Clinical Nutrition and Management, Graduate Division of Nutritional and Environmental Sciences, and Graduate School of Integrated Pharmaceutical and Nutritional Sciences, The University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Megumi Sato
- Laboratory of Clinical Nutrition and Management, Graduate Division of Nutritional and Environmental Sciences, and Graduate School of Integrated Pharmaceutical and Nutritional Sciences, The University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Toshio Hosaka
- Laboratory of Clinical Nutrition, Graduate Division of Nutritional and Environmental Sciences, and Graduate School of Integrated Pharmaceutical and Nutritional Sciences, The University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hidekazu Arai
- Laboratory of Clinical Nutrition and Management, Graduate Division of Nutritional and Environmental Sciences, and Graduate School of Integrated Pharmaceutical and Nutritional Sciences, The University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
37
|
Fajardo VC, Barreto SM, Coelho CG, Haueisen Sander Diniz MDF, Bisi Molina MDC, Pinho Ribeiro AL, Telles RW. Ultra-processed foods: Cross-sectional and longitudinal association with uric acid and hyperuricemia in ELSA-Brasil. Nutr Metab Cardiovasc Dis 2023; 33:75-83. [PMID: 36411223 DOI: 10.1016/j.numecd.2022.09.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/16/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND AIMS Food intake influences uric acid (UA) levels and hyperuricemia (HU), but evidence on the role of ultra-processed foods (UPFs) are scarce. The association between UPFs consumption and (1) HU prevalence and UA levels; (2) HU cumulative incidence; and (3) UA level change over a 4-year period was investigated. METHODS AND RESULTS Cross-sectional and longitudinal analyses were performed using baseline (2008-2010, aged 35-74 years) and second visit (2012-2014) data from the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil). Participants with glomerular filtration rate <60 mL/min/1.73 m2, bariatric surgery, implausible caloric intake, and using urate-lowering therapy (ULT) at baseline were excluded (all analyses). Participants with HU at baseline were excluded from longitudinal analyses. UPFs consumption was assessed using a food frequency questionnaire (FFQ) and categorized by the NOVA classification system (100 g/day). HU was defined as UA≥6.8 mg/dL. Linear, logistic, and mixed-effect linear regressions investigated the associations between UPFs consumption and UA/HU, adjusted for covariates. The final samples included 13,923 (cross-sectional) and 10,517 (longitudinal) individuals. The prevalence of HU was 18.7%, and the cumulative incidence was 4.9%. Greater UPFs consumption was associated with a greater prevalence of HU (OR:1.025 95%CI: 1.006; 1.044) and higher UA levels (β:0.024 95%CI: 0.016; 0.032). Every additional consumption of 100 g/day of UPFs raised the 4-year cumulative incidence of HU by 5.6% (95%CI: 1.021; 1.092). However, UPFs were not associated with the pace of UA level changes during the study period. CONCLUSION The present study shows that greater UPFs consumption is associated with another deleterious health consequence: higher UA levels and the risk of having HU.
Collapse
Affiliation(s)
- Virgínia Capistrano Fajardo
- Post-graduate Program in Ciências Aplicadas à Saúde do Adulto, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sandhi Maria Barreto
- Department of Preventive Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Hospital das Clínicas da UFMG-Ebserh, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carolina Gomes Coelho
- Department of Preventive Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Hospital das Clínicas da UFMG-Ebserh, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria de Fátima Haueisen Sander Diniz
- Hospital das Clínicas da UFMG-Ebserh, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Internal Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Antônio Luiz Pinho Ribeiro
- Department of Internal Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Telehealth Center, Hospital das Clínicas da UFMG-Ebserh, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rosa Weiss Telles
- Department of Internal Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Rheumatology Service, Hospital das Clínicas da UFMG-Ebserh, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
38
|
Wang P, Zhang X, Zheng X, Gao J, Shang M, Xu J, Liang H. Folic Acid Protects against Hyperuricemia in C57BL/6J Mice via Ameliorating Gut-Kidney Axis Dysfunction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:15787-15803. [PMID: 36473110 DOI: 10.1021/acs.jafc.2c06297] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Emerging lines of research evidence point to a vital role of gut-kidney axis in the development of hyperuricemia (HUA), which has been identified as an increasing burden worldwide due to the high prevalence. The involved crosstalk which links the metabolic and immune-related pathways is mainly responsible for maintaining the axial homeostasis of uric acid (UA) metabolism. Nowadays, the urate-lowering drugs only aim to treat acute gouty arthritis as a result of their controversial clinical application in HUA. In this study, we established the HUA model of C57BL/6J mice to evaluate the effectiveness of folic acid on UA metabolism and further explored the underlying mechanisms. Folic acid attenuated the kidney tissue injury and excretion dysfunction, as well as the typical fibrosis in HUA mice. Molecular docking results also revealed the structure-activity relationship of the folic acid metabolic unit and the UA transporters GLUT9 and URAT1, implying the potential interaction. Also, folic acid alleviated HUA-induced Th17/Treg imbalance and intestinal tissue damage and inhibited the active state of the TLR4/NF-κB signaling pathway, which is closely associated with the circulating LPS level caused by the impaired intestinal permeability. Furthermore, the changes of intestinal microecology induced by HUA were restored by folic acid, including the alteration in the structure and species composition of the gut microbiome community, and metabolite short-chain fatty acids. Collectively, this study revealed that folic acid intervention exerted improving effects on HUA by ameliorating gut-kidney axis dysfunction.
Collapse
Affiliation(s)
- Peng Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Ning Xia Road 308, Qingdao 266071, China
| | - Xiaoqi Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Ning Xia Road 308, Qingdao 266071, China
| | - Xian Zheng
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Ning Xia Road 308, Qingdao 266071, China
| | - Jingru Gao
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Ning Xia Road 308, Qingdao 266071, China
| | - Mengfei Shang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Ning Xia Road 308, Qingdao 266071, China
| | - Jinghan Xu
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Ning Xia Road 308, Qingdao 266071, China
| | - Hui Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Ning Xia Road 308, Qingdao 266071, China
| |
Collapse
|
39
|
Tong S, Zhang P, Cheng Q, Chen M, Chen X, Wang Z, Lu X, Wu H. The role of gut microbiota in gout: Is gut microbiota a potential target for gout treatment. Front Cell Infect Microbiol 2022; 12:1051682. [PMID: 36506033 PMCID: PMC9730829 DOI: 10.3389/fcimb.2022.1051682] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
Numerous studies have demonstrated that gut microbiota is essential for the host's health because it regulates the host's metabolism, endocrine, and immune systems. In recent years, increasing evidence has shown that gut microbiota plays a role in the onset and progression of gout. Changes in the composition and metabolism of the gut microbiota, result in abnormalities of uric acid degradation, increasing uric acid generation, releasing pro-inflammatory mediators, and intestinal barrier damage in developing gout. As a result, gout therapy that targets gut microbiota has drawn significant interest. This review summarized how the gut microbiota contributes to the pathophysiology of gout and how gout affects the gut microbiota. Additionally, this study explained how gut microbiota might serve as a unique index for the diagnosis of gout and how conventional gout treatment medicines interact with it. Finally, prospective therapeutic approaches focusing on gut microbiota for the prevention and treatment of gout were highlighted, which may represent a future avenue in gout treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoyong Lu
- *Correspondence: Xiaoyong Lu, ; Huaxiang Wu,
| | - Huaxiang Wu
- *Correspondence: Xiaoyong Lu, ; Huaxiang Wu,
| |
Collapse
|
40
|
Supplementation of Lactobacillus plantarum (TCI227) Prevented Potassium-Oxonate-Induced Hyperuricemia in Rats. Nutrients 2022; 14:nu14224832. [PMID: 36432519 PMCID: PMC9693167 DOI: 10.3390/nu14224832] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
Hyperuricemia (HC) is one of the important risk factors for gout, arteriosclerosis, and cardiovascular disease. Animal studies have shown that Lactobacillus plantarum can improve microbiota and immune regulation, as well as inhibit uric acid production. However, it is not clear whether L. plantarum can improve HC and intestinal microbiota. We used potassium oxonate (PO) to induce HC in male SD rats and then treated them with L. plantarum TCI227 in a dose-dependent manner (HC + LD, HC + MD, HC + HD) for 4 weeks. We examined organ weight, conducted biochemical examinations of blood and urine, and analyzed the intestinal microbiota in feces through a 16s rDNA sequence analysis. In this study, TCI227 improved body weight, decreased creatinine and serum uric acid, and increased urine uric acid compared to the HC group. Furthermore, TCI227 increased short-chain fatty acids (SCFAs). In the fecal microbiota (family), TCI227 increased the level of Lactobacillaceae and then decreased the levels of Deferribacteres and Prevotellaceae compared to the HC group. Finally, in the fecal microbiota (genus), TCI227 decreased the level of Prevotella and then increased the levels of Lactobacillus and Ruminococcus compared to the HC group. This study suggested that TCI227 can improve HC and can change the composition of intestinal microbiota in PO-induced male HC SD rats.
Collapse
|
41
|
Johnson RJ, Sánchez-Lozada LG, Nakagawa T, Rodriguez-Iturbe B, Tolan D, Gaucher EA, Andrews P, Lanaspa MA. Do thrifty genes exist? Revisiting uricase. Obesity (Silver Spring) 2022; 30:1917-1926. [PMID: 36150210 PMCID: PMC9512363 DOI: 10.1002/oby.23540] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/26/2022] [Accepted: 06/10/2022] [Indexed: 11/07/2022]
Abstract
Sixty years ago, the geneticist James Neel proposed that the epidemics of obesity and diabetes today may have evolutionary roots. Specifically, he suggested that our ancestors may have accumulated mutations during periods of famine that provided a survival advantage at that time. However, the presence of this "thrifty genotype" in today's world, where food is plentiful, would predispose us to obesity and diabetes. The "thrifty gene" hypothesis, attractive to some, has been challenged over the years. The authors have previously postulated that the loss of the uricase gene, resulting in a rise in serum and intracellular uric acid levels, satisfies the criteria of a thrifty genotype mutation. This paper reviews and brings up-to-date the evidence supporting the hypothesis and discusses the current arguments that challenge this hypothesis. Although further studies are needed to test the hypothesis, the evidence supporting a loss of uricase as a thrifty gene is substantial and supports a role for evolutionary biology in the pathogenesis of the current obesity and diabetes epidemics.
Collapse
Affiliation(s)
- Richard J Johnson
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | | | - Bernardo Rodriguez-Iturbe
- Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico and INC Ignacio Chavez, Mexico City, Mexico
| | - Dean Tolan
- Biology Department, Boston University, Boston MA
| | - Eric A. Gaucher
- Department of Biology, Georgia State University, Atlanta, GA
| | - Peter Andrews
- Department of Earth Sciences, Natural History Museum, London, UK
| | - Miguel A Lanaspa
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Nephrology, Oregon Health Sciences University
| |
Collapse
|
42
|
Lee Y, Kim N, Werlinger P, Suh DA, Lee H, Cho JH, Cheng J. Probiotic Characterization of Lactobacillus brevis MJM60390 and In Vivo Assessment of Its Antihyperuricemic Activity. J Med Food 2022; 25:367-380. [PMID: 35438552 DOI: 10.1089/jmf.2021.k.0171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Uric acid is the final product of purine metabolism in human. The increase of serum uric acid is tightly related to the incidence of hyperuricemia and gout. Also, it has been reported that the intake of purine-rich foods like meat and seafood is associated with an increased risk of gout. Therefore, the reduction of purine absorption is one of therapeutic approaches to prevent hyperuricemia and gout. Currently, probiotics are being studied for the management of hyperuricemia and gout. In this study, we aimed to investigate the effect of Lactobacillus brevis MJM60390 on hyperuricemia induced by a high-purine diet and potassium oxonate in a mouse model. L. brevis MJM60390 among 24 lactic acid bacteria isolated from fermented foods showed the highest ability to assimilate inosine and guanosine in vitro and typical probiotic characteristics, like the absence of bioamine production, D-lactate production, hemolytic activity, as well as tolerance to simulated orogastrointestinal conditions and adherence to Caco-2 cells. In an in vivo animal study, the uric acid level in serum was significantly reduced to a normal level after oral administration of L. brevis MJM60390 for 2 weeks. The activity of xanthine oxidase catalyzing the formation of uric acid was also inhibited by 30%. Interestingly, damage to the glomerulus, Bowman's capsule, and tubules in the hyperuricemia model were reversed by supplementation with this strain. Fecal microbiome analysis revealed that L. brevis MJM60390 supplementation enhanced the relative abundance of the Rikenellaceae family, which produces the short-chain fatty acid butyrate and helps to maintain good gut condition. Therefore, these results demonstrated that L. brevis MJM60390 can be a probiotic candidate to prevent hyperuricemia.
Collapse
Affiliation(s)
- Youjin Lee
- Graduate School of Interdisciplinary Program of Biomodulation, and Myongji University, Yongin-si, Gyeonggi-Do, Korea
| | - Neagawooridwimeu Kim
- Graduate School of Interdisciplinary Program of Biomodulation, and Myongji University, Yongin-si, Gyeonggi-Do, Korea
| | - Pia Werlinger
- Graduate School of Interdisciplinary Program of Biomodulation, and Myongji University, Yongin-si, Gyeonggi-Do, Korea
| | - Dong-A Suh
- Graduate School of Interdisciplinary Program of Biomodulation, and Myongji University, Yongin-si, Gyeonggi-Do, Korea
| | - Hanki Lee
- Graduate School of Interdisciplinary Program of Biomodulation, and Myongji University, Yongin-si, Gyeonggi-Do, Korea
| | - Joo-Hyung Cho
- Graduate School of Interdisciplinary Program of Biomodulation, and Myongji University, Yongin-si, Gyeonggi-Do, Korea
| | - Jinhua Cheng
- Graduate School of Interdisciplinary Program of Biomodulation, and Myongji University, Yongin-si, Gyeonggi-Do, Korea.,Myongji Bioefficacy Research Center, Myongji University, Yongin-si, Gyeonggi-Do, Korea
| |
Collapse
|
43
|
Gao Y, Yu Y, Qin W, Fan N, Qi Y, Chen H, Duan W. Uricase-deficient rats with similarly stable serum uric acid to human’s are sensitive model animals for studying hyperuricemia. PLoS One 2022; 17:e0264696. [PMID: 35239728 PMCID: PMC8893661 DOI: 10.1371/journal.pone.0264696] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
The aim of this study was to provide a sensitive model animal for studying hyperuricemia. Male uricase-deficient rats, named Kunming-DY rats, were raised for 130 days, or orally administered with purines and other chemicals. Serum uric acid (SUA) in the animals was assayed, and the UA level in their organs and their 24-h excretion was determined. Genes in the jejunum, ileum, kidney and liver related to UA synthesis and transportation were detected by quantitative RNA sequencing. Uricase-deficient rats have a high level of SUA and are sensitive to xanthine, adenosine, inosine, allopurinol, and alcohol. Besides, the high level of SUA in male uricase-deficient rats was stable, much higher than that in wild-type rats but similar to that in men. The distribution pattern of UA in uricase-deficient rats’ organs was different from that in wild-type rats. The kidney, liver, and small intestine were the top three organs where UA distributed, but the UA in the small intestine, colon, lung, thymus, and brain was less affected by uricase deficiency, indicating that these organs are constitutive distribution organs in UA. The 24-h UA excreted by a uricase-deficient rat was about five times higher than that excreted by a wild-type rat. However, the 24-h UA excreted through feces was not significantly changed. Both the urine volume and UA in uricase-deficient rats significantly increased, and more than 90% of UA was excreted via urine. The expression of xanthine dehydrogenase was not upregulated. Some genes of transporter associated with uric acid excretion in the kidney were significantly regulated, though not sufficient to explain the increase in SUA. In conclusion, male uricase-deficient rats’ UA metabolism is similar to that of men. The elevation of SUA in uricase-deficient rats is caused by uricase deficiency, and uricase-deficient rats are a sensitive model for studying hyperuricemia.
Collapse
Affiliation(s)
- Yinfang Gao
- The Department of Pharmacology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yun Yu
- The Department of Pharmacology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Wan Qin
- The Department of Pathology, School of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan Province, China
| | - Nan Fan
- The Department of Pharmacology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yalin Qi
- The Department of Pharmacology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Huan Chen
- The Department of Pharmacology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
| | - Weigang Duan
- The Department of Pharmacology, School of Basic Medicine, Kunming Medical University, Kunming, Yunnan Province, China
- * E-mail:
| |
Collapse
|
44
|
Song S, Lou Y, Mao Y, Wen X, Fan M, He Z, Shen Y, Wen C, Shao T. Alteration of Gut Microbiome and Correlated Amino Acid Metabolism Contribute to Hyperuricemia and Th17-Driven Inflammation in Uox-KO Mice. Front Immunol 2022; 13:804306. [PMID: 35197978 PMCID: PMC8858814 DOI: 10.3389/fimmu.2022.804306] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/12/2022] [Indexed: 12/11/2022] Open
Abstract
Although gut dysbiosis had been demonstrated to be an important factor affecting hyperuricemia (HUA) and gout, little is known for its potential mechanistic connections. In this study, Uox-KO mice model that with spontaneously developed pronounced HUA and urate nephropathy was used to explore the pathophysiologic mechanism of microbiota alterations in HUA and gout with integrated multi-omics analysis. 16S rRNA gene sequencing was performed to characterize the characteristic bacteria, and untargeted LC/MS analysis was applied to reveal the featured metabolites. Our results showed there was a significant shift in gut microbiota composition and function in Uox-KO mice compared to WT mice and apparent metabolomics differences between the two groups. Among them, amino acids metabolism appears to play a critical role. Correlation analysis further revealed that the characteristic metabolites were strongly influenced by the discrepant bacterial genera. Furthermore, impairment of intestinal integrity and profound alterations in the profile of solute carrier family resulted in dysregulation of amino acids transportation, which subsequently impacted serum uric acid level and CD4+ Th17 driven inflammation. Together, these data indicate that gut dysbiosis promotes purine metabolism disorder and inflammation in Uox-KO mice. Remodeling the gut microbiota is a promising strategy to combat HUA and gout.
Collapse
Affiliation(s)
- Siyue Song
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Lou
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yingying Mao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianghui Wen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Moqi Fan
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhixing He
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Shen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengping Wen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Chengping Wen, ; Tiejuan Shao,
| | - Tiejuan Shao
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Chengping Wen, ; Tiejuan Shao,
| |
Collapse
|
45
|
Serum Uric Acid Levels in Parkinson’s Disease: A Cross-Sectional Electronic Medical Record Database Study from a Tertiary Referral Centre in Romania. Medicina (B Aires) 2022; 58:medicina58020245. [PMID: 35208569 PMCID: PMC8877142 DOI: 10.3390/medicina58020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background and Objectives: Parkinson’s disease (PD) is a prevalent neurodegenerative condition responsible for progressive motor and non-motor symptoms. Currently, no prophylactic or disease-modifying interventions are available. Uric acid (UA) is a potent endogenous antioxidant, resulting from purine metabolism. It is responsible for about half of the antioxidant capacity of the plasma. Increasing evidence suggests that lower serum UA levels are associated with an increased risk of developing PD and with faster disease progression. Materials and Methods: We conducted an electronic medical record database study to investigate the associations between UA levels and different characteristics of PD. Results: Out of 274 datasets from distinct patients with PD, 49 complied with the predefined inclusion and exclusion criteria. Lower UA levels were significantly associated with the severity of parkinsonism according to the Hoehn and Yahr stage (rs = 0.488, p = 0.002), with the motor complications of long-term dopaminergic treatment (r = 0.333, p = 0.027), and with the presence of neurocognitive impairment (r = 0.346, p = 0.021). Conclusions: Oxidative stress is considered a key player in the etiopathogenesis of PD, therefore the involvement of lower UA levels in the development and progression of PD is plausible. Data on the potential therapeutic roles of elevating serum UA (e.g., by precursor administration or diet manipulation) are scarce, but considering the accumulating epidemiological evidence, the topic warrants further research.
Collapse
|
46
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:919-929. [DOI: 10.1093/jpp/rgac024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 04/03/2022] [Indexed: 11/14/2022]
|
47
|
Wang C, Dou X, Li J, Wu J, Cheng Y, An N. Composition and Diversity of the Ocular Surface Microbiota in Patients With Blepharitis in Northwestern China. Front Med (Lausanne) 2021; 8:768849. [PMID: 34950683 PMCID: PMC8688757 DOI: 10.3389/fmed.2021.768849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/17/2021] [Indexed: 12/27/2022] Open
Abstract
Purpose: To investigate the composition and diversity of the microbiota on the ocular surface of patients with blepharitis in northwestern China via 16S rDNA amplicon sequencing. Methods: Thirty-seven patients with blepharitis divided into groups of anterior, posterior and mixed blepharitis and twenty healthy controls from northwestern China were enrolled in the study. Samples were collected from the eyelid margin and conjunctival sac of each participant. The V3–V4 region of bacterial 16S rDNA in each sample was amplified and sequenced on the Illumina HiSeq 2500 sequencing platform, and the differences in taxonomy and diversity among different groups were compared. Results: The composition of the ocular surface microbiota of patients with blepharitis was similar to that of healthy subjects, but there were differences in the relative abundance of each bacterium. At the phylum level, the abundances of Actinobacteria, Cyanobacteria, Verrucomicrobia, Acidobacteria, Chloroflexi, and Atribacteria were significantly higher in the blepharitis group than in the healthy control group, while the relative abundance of Firmicutes was significantly lower (p < 0.05, Mann-Whitney U). At the genus level, the abundances of Lactobacillus, Ralstonia, Bacteroides, Akkermansia, Bifidobacterium, Escherichia-Shigella, Faecalibacterium, and Brevibacterium were significantly higher in the blepharitis group than in the healthy control group, while the relative abundances of Bacillus, Staphylococcus, Streptococcus, and Acinetobacter were significantly lower in the blepharitis group (p < 0.05, Mann-Whitney U). The microbiota of anterior blepharitis was similar to that of mixed blepharitis but different from that of posterior blepharitis. Lactobacillus and Bifidobacterium are biomarkers of posterior blepharitis, and Ralstonia is a biomarker of mixed blepharitis. There was no significant difference in the ocular surface microbiota between the eyelid margin and conjunctival sac with or without blepharitis. Conclusion: The ocular surface microbiota of patients with blepharitis varied among different study groups, according to 16S rDNA amplicon sequencing analysis. The reason might be due to the participants being from different environments and having different lifestyles. Lactobacillus, Bifidobacterium, Akkermansia, Ralstonia, and Bacteroides may play important roles in the pathogenesis of blepharitis.
Collapse
Affiliation(s)
- Changhao Wang
- College of Life Science, Northwest University, Xi'an, China
| | - Xiuhong Dou
- College of Life Science, Northwest University, Xi'an, China
| | - Jian Li
- College of Life Science, Northwest University, Xi'an, China
| | - Jie Wu
- Department of Ophthalmology, Xi'an No.1 Hospital, First Affiliated Hospital of Northwest University, Xi'an, China.,Shaanxi Key Laboratory of Ophthalmology, Shaanxi Provincial Clinical Research Center for Ophthalmic Diseases, Shaanxi Institute of Ophthalmology, Xi'an, China
| | - Yan Cheng
- Department of Ophthalmology, Xi'an No.1 Hospital, First Affiliated Hospital of Northwest University, Xi'an, China.,Shaanxi Key Laboratory of Ophthalmology, Shaanxi Provincial Clinical Research Center for Ophthalmic Diseases, Shaanxi Institute of Ophthalmology, Xi'an, China
| | - Na An
- Department of Ophthalmology, Xi'an No.1 Hospital, First Affiliated Hospital of Northwest University, Xi'an, China.,Shaanxi Key Laboratory of Ophthalmology, Shaanxi Provincial Clinical Research Center for Ophthalmic Diseases, Shaanxi Institute of Ophthalmology, Xi'an, China
| |
Collapse
|
48
|
Lin G, Yu Q, Xu L, Huang Z, Mai L, Jiang L, Su Z, Xie J, Li Y, Liu Y, Lin Z, Chen J. Berberrubine attenuates potassium oxonate- and hypoxanthine-induced hyperuricemia by regulating urate transporters and JAK2/STAT3 signaling pathway. Eur J Pharmacol 2021; 912:174592. [PMID: 34699754 DOI: 10.1016/j.ejphar.2021.174592] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/05/2021] [Accepted: 10/20/2021] [Indexed: 01/27/2023]
Abstract
Phellodendri Chinensis Cortex (PC) is a traditional medicinal material used to treat gout and hyperuricemia (HUA) in China. Berberine (BBR), the main component of PC, possesses anti-hyperuricemic and anti-gout effects. However, BBR exhibits low bioavailability due to its extensive metabolism and limited absorption. Thus, the metabolites of BBR are believed to be the potential active forms responsible for its in vivo biological activities. Berberrubine (BRB), one of the major metabolites of BBR, exhibits appreciable biological activities even superior to BBR. In this work, the anti-hyperuricemic efficacy of BRB was investigated in HUA model mice induced by co-administration with intraperitoneal potassium oxonate (PO) and oral hypoxanthine (HX) for 7 days. Results showed that administration with BRB (6.25, 12.5, and 25.0 mg/kg) significantly decreased the serum levels of uric acid (UA) by 49.70%, 75.35%, and 75.96% respectively, when compared to the HUA group. In addition, BRB sharply decreased the levels of blood urea nitrogen (BUN) (by 19.62%, 28.98%, and 38.72%, respectively) and serum creatinine (CRE) (by 16.19%, 25.07%, and 52.08%, respectively) and reversed the PO/HX-induced renal histopathological damage dose-dependently. Additionally, BRB lowered the hepatic XOD activity, downregulated the expressions of glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1), upregulated expressions of organic anion transporter 1/3 (OAT1/3) and ATP-binding cassette transporter subfamily G member 2 (ABCG2) at both protein and mRNA levels, and suppressed the activation of the JAK2/STAT3 signaling pathway. In addition, BRB significantly decreased the levels of inflammatory mediators (IL-1β, IL-6, and TNF-α). In conclusion, our study indicated that BRB exerted anti-hyperuricemic effect, at least in part, via regulating the urate transporter expressions and suppressing the JAK2/STAT3 signaling pathway. BRB was believed to be promising for further development into a potential therapeutic agent for HUA treatment.
Collapse
Affiliation(s)
- Guoshu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China
| | - Qiuxia Yu
- The Second Clinical College of Guangzhou University of Chinese Medicine, 510120, Guangzhou, P.R. China
| | - Lieqiang Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China
| | - Ziwei Huang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, 510405, Guangzhou, P.R. China
| | - Liting Mai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China
| | - Linyun Jiang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, 510405, Guangzhou, P.R. China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, P.R. China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 510120, Guangzhou, P.R. China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, 510120, Guangzhou, P.R. China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China
| | - Zhixiu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China.
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, 510006, Guangzhou, P.R. China.
| |
Collapse
|
49
|
Yang HT, Xiu WJ, Liu JK, Yang Y, Hou XG, Zheng YY, Wu TT, Wu CX, Xie X. Gut Microbiota Characterization in Patients with Asymptomatic Hyperuricemia: probiotics increased. Bioengineered 2021; 12:7263-7275. [PMID: 34590550 PMCID: PMC8806635 DOI: 10.1080/21655979.2021.1976897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/31/2021] [Indexed: 10/27/2022] Open
Abstract
Asymptomatic hyperuricemia (AH) is an early stage of gout. Emerging evidence shows that the intestinal microbiota is related to gout. However, the relationship between AH and the intestinal microbiota is poorly understood. Therefore, the aim of the current study was to explore the possible correlation between AH and intestinal flora. We compared the intestinal microbial communities of AH (45 cases) and healthy subjects (45 cases) by 16S rRNA gene sequencing and clustering analysis on the incorporated population. Intestinal-type clustering can be divided into two groups, and significant differences in the proportion of AH are found among different bowel types. Alpha diversity indices were higher in the AH group than in the control group, and beta diversity indices also showed significant differences. A total of 19 genera were found different between the AH group and the control group. Compared with the control group, some probiotics are increased in the AH population. Two groups were ranked by importance of bacteria. We found the different bacteria partially coincided with the important bacteria, and the joint diagnosis level of the important bacteria was good. Conclusion: There were significant differences in the composition of intestinal biota between AH patients and healthy subjects. Some probiotics increased in AH.
Collapse
Affiliation(s)
- Hai-Tao Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wen-Juan Xiu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jing-Kun Liu
- Department of Oncology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yi Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chen-Xin Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
50
|
Li X, Gao X, Zhang H, Liu Y, Sarker MMR, Wu Y, Chen X, Zhao C. The anti-hyperuricemic effects of green alga Enteromorpha prolifera polysaccharide via regulation of the uric acid transporters in vivo. Food Chem Toxicol 2021; 158:112630. [PMID: 34687833 DOI: 10.1016/j.fct.2021.112630] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 11/15/2022]
Abstract
A novel polysaccharide obtained from Enteromorpha prolifera (EPP) was purified through diethylaminoethyl cellulose-52 and Sephadex G-75 chromatography. Fourier transform infrared spectroscopy, high-performance liquid chromatography, and nuclear magnetic resonance (NMR) spectroscopy were employed to analyse the structure of EPP. It mainly comprised rhamnose, glucuronic acid, galactose, arabinose, and xylose at a molar ratio of 20.45:12.74:10.99:5.84:1.95, and its average molecular weight was 46.56 kDa. The seven major glycosidic residues identified by NMR were as follows: →2)-α-L-Araf-(1→, →2)-α-L-Rhap-(1→, →4)-α-L-Rhap-(1→, →2,6)-β-D-Galp-(1→, →4)-β-D-GlcpA-(1→, →3,4)-β-D-GlcpA-(1→, and →4)-β-Xylp-(1→. The effect of EPP on hyperuricemic mice was determined by analysing correlative general physical parameters, renal histopathology, renal gene expressions, and gut microbiome. EPP significantly reduced serum uric acid (UA), serum blood urea nitrogen, serum xanthine oxidase (XOD), and hepatic XOD as well as improved histological parameters in hyperuricemic mice. Furthermore, mRNA and protein expression analyses showed the upregulation of UA excretion genes such as ABCG2, OAT1, and NPT1 and downregulation of UA resorption gene URAT1. Moreover, EPP maintained the stability of the intestinal flora and confirmed that Parasutterella is closely related to the regulation of hyperuricemia. This study is the first to demonstrate the anti-hyperuricemic activity of EPP and highlight its therapeutic potential for hyperuricemia-related diseases.
Collapse
Affiliation(s)
- Xiaoqing Li
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiaoxiang Gao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Hui Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yuanyuan Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Md Moklesur Rahman Sarker
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road, Dhanmondi, Dhaka, 1205, Bangladesh; Pharmacology and Toxicology Research Division, Health Med Science Research Limited, 3/1 Lalmatia, 1207, Dhaka, Bangladesh
| | - Yijing Wu
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China.
| | - Xinhua Chen
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Chao Zhao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|