1
|
Gao S, Xiao AY, Zou S, Li T, Deng H, Wang Y. Exploring causal links in the gut-brain axis: a Mendelian randomization study of gut microbiota, metabolites, and cognition. Food Funct 2025. [PMID: 40423497 DOI: 10.1039/d4fo04366a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
The causal mediation effects of metabolites between gut microbiota and cognitive phenotypes remain unclear. Guided by the gut-brain axis mechanism, this study employed systematic Mendelian randomization (MR) to investigate these mediation pathways and their implications for functional food development. Univariate MR analysis was performed to estimate the causality of 211 gut microbial taxa (n = 18 340) and 452 serum metabolites (n = 7824) on general cognitive (n = 257 700), non-cognitive (n = 510 795), and specific cognitive phenotypes (n ≈ 2500) using genome-wide association study data. Inverse-variance weighted estimation was adopted as the primary method, with MR sensitivity analyses performed to complement the results. Metabolic pathway analysis was employed to enrich metabolic profiles, while two-step MR was used to screen mediation pathways. We revealed seven causal associations between microbiotas or metabolites and cognitive phenotypes (FDR < 0.05). Increased abundance of the order Clostridiales id.1863 was associated with better cognitive traits (OR = 1.14, 95%CI = 1.06-1.22, P = 2.06 × 10-4), while 1-linoleoylglycerophosphoethanolamine was also positively associated with cognitive traits (OR = 1.61, 95%CI = 1.33-1.95, P = 8.17 × 10-7). Seven significant metabolic pathways were enriched, including alpha-linolenic acid and linoleic acid metabolism, highlighting the potential role of omega-3 and omega-6 fatty acids in cognitive health. We further identified two significant mediation pathways linking the gut microbiota to cognitive phenotypes through metabolites. Notably, homostachydrine (39.1%) was found to mediate a proportion of the impact of the genus Turicibacter on emotion recognition (indirect effect: β = 0.105, 95%CI = 0.006-0.259, p = 2.60 × 10-2). This study provides evidence for causal relationships between gut microbiota, serum metabolites, and cognitive function, supporting the gut-brain axis mechanism. Our findings suggest potential targets for the development of functional food and personalized nutrition to improve cognitive health.
Collapse
Affiliation(s)
- Sunan Gao
- School of Statistics, Renmin University of China, Beijing, China
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, USA
| | - Angela Y Xiao
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Siyu Zou
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, USA
- School of Public Health, Peking University, Beijing, China
| | - Tongxu Li
- School of Statistics, Renmin University of China, Beijing, China
| | - Heming Deng
- School of Statistics, Renmin University of China, Beijing, China
| | - Yu Wang
- Center for Applied Statistics, Renmin University of China, Beijing, China.
- School of Statistics, Renmin University of China, Beijing, China
| |
Collapse
|
2
|
Liu P, Hu P, Jin M, Sun W, Wu J, Tang Y, Shi D, Xie T, Tong Y, Huang L, Zhang D, Zheng H, Xu X, He H. Compound probiotics alleviate hyperuricemia-induced renal injury via restoring gut microbiota and metabolism. BMC Microbiol 2025; 25:280. [PMID: 40335932 PMCID: PMC12060558 DOI: 10.1186/s12866-025-04012-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
To investigate the role and mechanisms of gut microbiota in hyperuricemia-induced renal injury, we established renal failure models using unilateral nephrectomized mice. After four weeks of adenine and potassium oxalate-supplemented diet, probiotic intervention was administered. Renal pathological and functional changes were assessed through H&E staining and plasma biochemical analysis. Gut microbiota composition and metabolite profiles were evaluated using 16 S rRNA gene sequencing and non-targeted metabolomics of fecal samples.Our findings demonstrate that the compound probioticS effectively attenuated hyperuricemia-associated renal dysfunction and interstitial fibrosis. The intervention reduced oxidative stress, mitophagy, and apoptosis in renal tubules. Probiotic treatment enhanced gut microbiota diversity, notably increasing the abundance of Prevotella_9, Dorea, and unclassified Bacteroidota, while decreasing unclassified Desulfovibrio. KEGG enrichment analysis revealed that probiotic intervention upregulated arginine and proline metabolism, as well as tyrosine metabolism in feces. Furthermore, it enhanced the metabolism of arginine, proline, valine, leucine, and isoleucine in plasma.Notably, sulfocholic acid and urocanic acid showed negative correlations with oxidative stress markers, autophagy, and apoptosis indicators. Similarly, plasma L-proline levels were inversely correlated with these pathological parameters.These results suggest that the compound probiotics may mitigate hyperuricemia-induced kidney damage through restoration of gut microbiota homeostasis and preservation of plasma and fecal metabolites. The protective mechanisms likely involve attenuation of hyperuricemia-associated oxidative stress, mitochondrial dysregulation, and phagocytosis-induced apoptosis.Our study provides compelling evidence that probiotic supplementation represents a promising therapeutic strategy for hyperuricemia-induced renal injury, potentially through modulation of gut microbiota and associated metabolic pathways.
Collapse
Affiliation(s)
- Ping Liu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Ping Hu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Meiping Jin
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Weiqian Sun
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Jiajun Wu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Yuyan Tang
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Danye Shi
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Ting Xie
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Yijing Tong
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Lusheng Huang
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Dongliang Zhang
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Hui Zheng
- Division of Endocrinology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| | - Xudong Xu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| | - Haidong He
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| |
Collapse
|
3
|
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
|
4
|
Han R, Li Y, Guo Y, Ren M, Shan M, Mao T, Qi X, Li Y, Tian Z, Fu T. Alginate ameliorates hyperuricemia in mice by restoring hyperuricemia-induced renal and intestinal dysfunctions. Int J Biol Macromol 2025; 310:143422. [PMID: 40268003 DOI: 10.1016/j.ijbiomac.2025.143422] [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: 01/17/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
Alginate, a bioactive polysaccharide fermentable by gut microbiota, has been shown to effectively reduce serum uric acid levels. However, its mechanisms and the role of gut microbiota remain unclear. In this study, we explored the effects of alginate with two different molecular weights on hyperuricemia mice. Both alginates exhibited potent hypouricemic effects through ABCG2 transporter upregulation, effectively ameliorating hyperuricemia-induced renal and intestinal dysfunctions, with the low-molecular-weight alginate demonstrating enhanced bioavailability through microbial biodegradation and superior therapeutic efficacy in hyperuricemia management. Additionally, we found that alginate alleviates gut microbiota dysbiosis induced by hyperuricemia by enriching potentially beneficial bacteria. These include Limosilactobacillus and Lactobacillus, which show a significant negative correlation with serum uric acid levels. These bacteria might regulate uric acid precursors during purine metabolism, thereby reducing uric acid accumulation. In summary, this study reveals the protective effects of alginate on renal and intestinal damage in hyperuricemia mice and highlights the crucial role of gut microbiota. It provides valuable insights into the mechanisms by which gut microbiota mediate the effects of alginate.
Collapse
Affiliation(s)
- Rongshuang Han
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China
| | - Yukun Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China
| | - Yingjie Guo
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China
| | - Minghan Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China
| | - Mengchen Shan
- Department of Gastroenterology, Qingdao Cardiovascular Hospital, Qingdao 266000, PR China
| | - Tao Mao
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China
| | - Xingsi Qi
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China
| | - Yupeng Li
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China.
| | - Tianyu Fu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266001, PR China.
| |
Collapse
|
5
|
Hou X, Wang M, Hu T, Wu Z, Liang H, Zhong Y, Ma Z, Zhang H, Xiao L, Zhang W, Zou Y. Evaluation of the safety and probiotic properties of Limosilactobacillus fermentum BGI-AF16, a uric acid-lowering probiotic strain. Microb Pathog 2025; 201:107382. [PMID: 39961375 DOI: 10.1016/j.micpath.2025.107382] [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: 05/06/2024] [Revised: 01/08/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
Some beneficial microorganisms in the intestine have the potential to degrade uric acid, offering a novel strategy for the prevention of hyperuricemia. In this study, the safety and probiotic potentials of Limosilactobacillus fermentum BGI-AF16 were evaluated by whole genome sequence analysis and in vitro experiments. Based on the gene analysis of antibiotic resistance and virulence factors, L. fermentum BGI-AF16 has been shown to be safe. We identified probiotic-related genes by genome annotation tools and conducted in vitro experiments to evaluate the ability of L. fermentum BGI-AF16 to inhibit pathogenic bacteria, tolerate a simulated gastrointestinal environment, and degrade uric acid. The results from in vitro experiments showed that L. fermentum BGI-AF16 had inhibitory effects on four clinically relevant pathogens and was highly tolerant to the gastrointestinal environment. In addition, L. fermentum BGI-AF16 was able to rapidly degrade uric acid within the first hour, and the strain could degrade 56.36 ± 2.32 % of uric acid by the third hour. The genome of the strain contains genes encoding flavin adenine dinucleotide (FAD)-dependent urate hydroxylase (EC.1.14.13.113), an enzyme that directly metabolizes uric acid. And the strain has a complete uric acid metabolic pathway. These results suggest that L. fermentum BGI-AF16 is a probiotic candidate with significant potential for reducing uric acid level.
Collapse
Affiliation(s)
- Xiaoxue Hou
- BGI Research, Shenzhen, 518083, China; College of Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Mengmeng Wang
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Zhinan Wu
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Yiyi Zhong
- BGI Precision Nutrition, Shenzhen, 518083, China
| | - Zhihui Ma
- BGI Precision Nutrition, Shenzhen, 518083, China
| | | | - Liang Xiao
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China
| | - Wenjin Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; BGI Genomics, Shenzhen, 518083, China
| | - Yuanqiang Zou
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China.
| |
Collapse
|
6
|
Logan AC, Mishra P. Aggression and Justice Involvement: Does Uric Acid Play a Role? Brain Sci 2025; 15:268. [PMID: 40149789 PMCID: PMC11940041 DOI: 10.3390/brainsci15030268] [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: 01/28/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
The search for biological markers that can be reliably linked to aggression and antisocial behavior has been central to the work of biological criminology. One such marker, uric acid, has long been suspected to play a causative role in promoting anger, irritability, aggression, and violence. Here, in this perspective article, we revisit some of the historical interest in uric acid as a compound relevant to brain and behavior, and reflect these early accounts off emergent scientific research. Advances in brain sciences, including neuropsychiatry and neuromicrobiology, have allowed for a more sophisticated understanding of potential mechanistic pathways linking uric acid with cognition and behavior. The updated science suggests that some of the early ideas surrounding uric acid and criminology had credibility. The available research strongly suggests that uric acid, as a potential biomarker of risk, is worthy of further research and close scrutiny. Informed by emergent gut-brain-microbiome research, we argue that certain aspects of early-to-mid-20th-century biological criminology were prematurely abandoned. From a legalome perspective, further advances surrounding uric acid and other gut-brain biomarkers can aid in shaping more humane, scientifically grounded policies that recognize the interplay between biology and environment.
Collapse
Affiliation(s)
| | - Pragya Mishra
- Department of Law, Central University of Allahabad, Prayagraj 211002, India;
| |
Collapse
|
7
|
Dong L, Dong F, Guo P, Li T, Fang Y, Dong Y, Xu X, Cai T, Liang S, Song X, Li L, Sun W, Zheng Y. Gut microbiota as a new target for hyperuricemia: A perspective from natural plant products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156402. [PMID: 39874797 DOI: 10.1016/j.phymed.2025.156402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/29/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Hyperuricemia, a prevalent chronic metabolic disorder caused by purine metabolism disturbances, is characterized by elevated serum uric acid (UA) levels. Prolonged hyperuricemia can cause severe complications such as gout or kidney damage. However, the toxic side effects of and adverse reactions to UA-lowering drugs are becoming increasingly prominent. Therefore, new targets and drugs for hyperuricemia are needed. PURPOSE This review aims to summarize recent research progress on the prevention and treatment mechanisms for gut microbiota-hyperuricemia from the perspective of plant-derived natural products. METHODS Data from PubMed, Web of Science, ScienceDirect, and the CNKI databases spanning from January 2020 to December 2024 were reviewed. The aim of this study is to categorize and summarize the relevant mechanisms through which natural products improve hyperuricemia via the gut microbiota. The retrieved data followed PRISMA criteria (Preferred Reporting Items for Systematic reviews and Meta-Analyses). RESULTS Regulating gut microbiota as a treatment for hyperuricemia. Targeting the gut microbiota could reduce host UA levels by promoting purine degradation, reducing UA production, and increasing UA excretion. Moreover, the gut microbiota also exerts anti-inflammatory and antioxidant effects that alleviate complications such as renal damage caused by hyperuricemia. Due to their diverse sources, multicomponent synergy, multitarget effects, and minimal side effects, plant-derived natural products have been extensively utilized in the management of hyperuricemia. Especially, utilizing natural products from plants to regulate the gut microbiota has become a new strategy for reducing UA levels. CONCLUSION This review comprehensively summarizes recent advances in understanding the preventive and therapeutic mechanisms of plant-derived natural products in ameliorating hyperuricemia and its comorbidities through gut microbiota modulation. This review contributes a novel perspective for the development of safer and more efficacious UA-lowering products.
Collapse
Affiliation(s)
- Ling Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Fengying Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Pingping Guo
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Tianxing Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China; Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100007, China
| | - Yini Fang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China; Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yang Dong
- Monitoring and Statistical Research Center, National Administration of Traditional Chinese Medicine, Beijing, 100021, China
| | - Xiaoxue Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Tianqi Cai
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Shufei Liang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Xinhua Song
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Lingru Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China.
| | - Wenlong Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China.
| | - Yanfei Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China.
| |
Collapse
|
8
|
Han QQ, Ren QD, Guo X, Farag MA, Zhang YH, Zhang MQ, Chen YY, Sun ST, Sun JY, Li NY, Liu C. Punicalagin attenuates hyperuricemia via restoring hyperuricemia-induced renal and intestinal dysfunctions. J Adv Res 2025; 69:449-461. [PMID: 38609050 PMCID: PMC11954802 DOI: 10.1016/j.jare.2024.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
INTRODUCTION It is estimated that 90% of hyperuricemia cases are attributed to the inability to excrete uric acid (UA). The two main organs in charge of excreting UA are the kidney (70%) and intestine (30%). Previous studies have reported that punicalagin (PU) could protect against kidney and intestinal damages, which makes it a potential candidate for alleviating hyperuricemia. However, the effects and deeper action mechanisms of PU for managing hyperuricemia are still unknown. OBJECTIVE To investigate the effect and action mechanisms of PU for ameliorating hyperuricemia. METHODS The effects and action mechanisms of PU on hyperuricemia were assessed using a hyperuricemia mice model. Phenotypic parameters, metabolomics analysis, and 16S rRNA sequencing were applied to explore the effect and fundamental action mechanisms inside the kidney and intestine of PU for improving hyperuricemia. RESULTS PU administration significantly decreased elevated serum uric acid (SUA) levels in hyperuricemia mice, and effectively alleviated the kidney and intestinal damage caused by hyperuricemia. In the kidney, PU down-regulated the expression of UA resorption protein URAT1 and GLUT9, while up-regulating the expression of UA excretion protein ABCG2 and OAT1 as mediated via the activation of MAKP/NF-κB in hyperuricemia mice. Additionally, PU attenuated renal glycometabolism disorder, which contributed to improving kidney dysfunction and inflammation. Similarly, PU increased UA excretion protein expression via inhibiting MAKP/NF-κB activation in the intestine of hyperuricemia mice. Furthermore, PU restored gut microbiota dysbiosis in hyperuricemia mice. CONCLUSION This research revealed the ameliorating impacts of PU on hyperuricemia by restoring kidney and intestine damage in hyperuricemia mice, and to be considered for the development of nutraceuticals used as UA-lowering agent.
Collapse
Affiliation(s)
- Qing-Qing Han
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150000, China
| | - Qi-Dong Ren
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China.
| | - Xu Guo
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Mohamed A Farag
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Yu-Hong Zhang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150000, China
| | - Meng-Qi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Ying-Ying Chen
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Shu-Tao Sun
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Jin-Yue Sun
- School of Public Health, Shandong Second Medical University, Weifang 261053, China.
| | - Ning-Yang Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P.R. China.
| | - Chao Liu
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs/Key Laboratory of Agro-Products Processing Technology of Shandong Province/Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China.
| |
Collapse
|
9
|
Zhang Y, Xu Y, Hu L, Wang X. Advancements related to probiotics for preventing and treating recurrent respiratory tract infections in children. Front Pediatr 2025; 13:1508613. [PMID: 39981209 PMCID: PMC11839809 DOI: 10.3389/fped.2025.1508613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Recurrent respiratory tract infections (RRTIs) are a common condition in pediatrics and significantly impact children's quality of life; however, their pathogenesis and contributing factors are not yet fully elucidated. Probiotics have recently emerged as promising agents for modulating intestinal microecology and have gained considerable attention in clinical research on preventing and treating RRTIs in children. This article provides an initial overview of the concept, classification, and mechanisms underlying probiotics. It emphasizes their beneficial effects on respiratory health by modulating intestinal microbial equilibrium, augmenting immune system functionality, and attenuating inflammatory responses. Subsequently, we examine existing research regarding the use of probiotics in pediatric RRTIs. Numerous clinical trials have unequivocally demonstrated that supplementing with probiotics can significantly reduce both the frequency and severity of RRTIs in children while also simultaneously decreasing antibiotic usage. However, there are ongoing controversies and challenges in current research concerning the influence of probiotic type, dosage, duration of use, and other factors on efficacy. Furthermore, variations have been observed across different studies. Additionally, it is crucial to further evaluate the safety and potential long-term side effects associated with probiotic use in children with RRTIs. In conclusion, we propose future research directions including conducting more high-quality randomized controlled trials to optimize application strategies for probiotics alongside other treatments while considering variations based on age and health conditions among pediatric populations. Finally, in summary although probiotics exhibit promising benefits in preventing and treating RRTIs in children; additional studies are necessary to refine their application strategies ensuring both safety and effectiveness.
Collapse
Affiliation(s)
- Yali Zhang
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yingying Xu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Ling Hu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xiaomei Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
10
|
Lv W, Chen H, Zhou P, Du A, Lei Y. Mechanisms Associated With Renal Injury in Hyperuricemia and Strategies for the Development of Natural Active Substances. Int J Rheum Dis 2025; 28:e70096. [PMID: 39895275 DOI: 10.1111/1756-185x.70096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
Hyperuricemia (HUA) is a metabolic condition resulting from an abnormality in the process of purine metabolism. Its occurrence has been on the rise globally. The results of relevant studies show that 5% to 12% of HUA patients will eventually develop gout, and one-third of these patients may involve the kidneys and develop kidney disease. Although the severe renal health hazards associated with excessive uric acid levels are well known, the specific molecular mechanisms remain unknown. Therefore, this paper provides insights into the mechanisms and related chain reactions of HUA leading to renal injury from three perspectives: imbalance of intestinal homeostasis, oxidative stress response, and NLRP3 inflammasome. In addition, standing against the background of the strong side effects and high tolerability disadvantages of commercially available uric acid-lowering drugs such as allopurinol, benzbromarone, and febuxostat, the development of a new active anti-hyperuricemic drug with fewer side effects is justified. This article reviews the progress of research on natural actives (probiotics, dietary polyphenols, peptides) with a high safety profile, multi-targeting, and integrative modulatory effects, in an attempt to provide some ideas for drug developers.
Collapse
Affiliation(s)
- Wanping Lv
- Outpatient Department, Chengdu Rheumatology Hospital, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huixiang Chen
- Hospitalization Department, Zhengzhou Gout and Rheumatology Hospital, Zhengzhou, China
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Pan Zhou
- Outpatient Department, Chengdu Rheumatology Hospital, Chengdu, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Aihua Du
- Hospitalization Department, Zhengzhou Gout and Rheumatology Hospital, Zhengzhou, China
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Yu Lei
- Outpatient Department, Chengdu Rheumatology Hospital, Chengdu, China
- School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
11
|
Xie R, Yuen SK, Tsang Z, Tai WCS, Yap DYH. The relationship between probiotics and prebiotics, kidney dysfunction and mortality - Results from a longitudinal cohort study and Mendelian randomization. Clin Nutr ESPEN 2025; 65:272-281. [PMID: 39672381 DOI: 10.1016/j.clnesp.2024.11.035] [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: 07/04/2024] [Revised: 11/08/2024] [Accepted: 11/29/2024] [Indexed: 12/15/2024]
Abstract
INTRODUCTION The benefits of probiotics/prebiotics consumption on chronic kidney disease (CKD) and mortality remains controversial. OBJECTIVES This study investigates the association of probiotics/prebiotics consumption with chronic kidney disease (CKD) and mortality. METHODS Clinical data were retrieved from the National Health and Nutrition Examination Survey (NHANES) 2005-2016 database. Weighted multivariable logistic and liner regression models, cox proportional hazards models and stratified analysis were used to analyse the relationships between consumption of probiotics/prebiotics, renal parameters, CKD and mortality. We also conducted a two-sample Mendelian randomization (MR) analysis of single nucleotide polymorphisms (SNPs) related to different genera of gut microbiota to assess their causal relationships with CKD and mortality. RESULTS 15,291 subjects were analysed (897 with consumption of probiotics/prebiotics and 14,394 without). The use of probiotics/prebiotics showed an inverse correlation with urinary albumin-to-creatinine ratio (UACR) (P < 0.05). Probiotics/prebiotics use was associated with lower risk of CKD in subjects with hypertension, hyperlipidaemia and diabetes mellitus. The consumption of probiotics/prebiotics was associated with a significantly lower risk of all-cause mortality in different regression models (P < 0.001, for all), but the lower risk of cardiovascular mortality did not reach statistical significance (P > 0.05, for all)]. MR analysis showed negative associations between the genetically predicted genus Flavonifractor and risk of CKD and diabetic kidney disease (DKD). CONCLUSION After multivariable regression, and cox proportional hazards analysis, we found that the use of probiotics/prebiotics was associated with improved kidney and mortality outcomes in the general population from NHANES database. The two-sample MR analysis provided further genetic evidence that a distinct genus of gut microbiota was associated with reduced risk of CKD, DKD and mortality.
Collapse
Affiliation(s)
- Ruiyan Xie
- Division of Nephrology, Department of Medicine, School of Clinical Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Sze Kit Yuen
- Renal Unit, Department of Medicine & Geriatrics, Caritas Medical Centre, Hong Kong
| | - Zoe Tsang
- Renal Unit, Department of Medicine & Geriatrics, Caritas Medical Centre, Hong Kong
| | - William C S Tai
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong; Laboratory for Probiotics and Prebiotics in Human Health, The Hong Kong Polytechnic University, Hong Kong
| | - Desmond Y H Yap
- Division of Nephrology, Department of Medicine, School of Clinical Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong.
| |
Collapse
|
12
|
Gu W, Zhao J, Xu Y. Hyperuricemia-induced complications: dysfunctional macrophages serve as a potential bridge. Front Immunol 2025; 16:1512093. [PMID: 39935474 PMCID: PMC11810932 DOI: 10.3389/fimmu.2025.1512093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
With the changes in modern life, hyperuricemia (HUA) has become a serious universal health issue, leading to rising morbidity and mortality. Characterized by elevated levels of UA, HUA has become an independent risk factor for gout, chronic kidney disease, insulin resistance, cardiovascular disease, nonalcoholic fatty liver disease, etc. As HUA is a metabolic syndrome, the immune response is likely to play an active role throughout the whole process. Moreover, macrophages, as an indispensable component of the immune system, may serve as a promising target for addressing hyperuricemia-induced inflammation. Along with their precursor cells, monocytes, macrophages play a key role in the pathogenesis of HUA, primarily through three specific aspects, all of which are associated with inflammatory cytokines. The first mechanism involves direct action on urate transporters, such as URAT1 and ABCG2. The second mechanism is the modulation of inflammation, including targeting toll-like receptors (TLRs) and the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome. The third mechanism pertains to the effects on oxidative stress mediators. In this review, we summarize the underlying mechanisms of hyperuricemia, focusing on the effects of macrophages, therapeutic approaches, and clinical trials addressing hyperuricemia-caused dysfunction. Additionally, we highlight directions for future development, aiming to support future theoretical studies.
Collapse
Affiliation(s)
- Wenyi Gu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajing Zhao
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yu Xu
- Department of Traditional Chinese Medicine, Shanghai Putuo Hospital of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for Traditional Chinese Medicine New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Wu J, Wang X, Aga L, Tang L, Tan S, Zhang D, Li H, Yang L, Zhang N, Su S, Xiao M, Min R, Li A, Wang X. Lacticaseibacillus casei JS-2 from 'Jiangshui' Reduces Uric Acid and Modulates Gut Microbiota in Hyperuricemia. Foods 2025; 14:407. [PMID: 39942000 PMCID: PMC11817023 DOI: 10.3390/foods14030407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Lacticaseibacillus casei (JS-2) is a novel probiotic isolated from "Jiangshui", a kind of traditional folk fermented food, which has a significant effect on hyperuricemia (HUA). In vitro experimental results showed that JS-2 has a high degradation ability and selectivity for uric acid (UA). The animal test results indicated that after two weeks of treatment, JS-2 could significantly reduce the level of UA in the serum of HUA quails (p < 0.01), and its effect is almost equivalent to that of the positive drug control group, benzbromarone. Further, after JS-2 treatment, the level of xanthine oxidase in quail serum decreased significantly. Analysis data of quail fecal metabolomics results showed that JS-2-altering metabolites were involved in amino acid, purine, and lipid metabolism. To investigate the mechanism underlying JS-2-mediated UA degradation in the quail model of HUA, 16S rRNA gene sequencing was conducted. It was found that the structure and function of the gut microbiota were restored after JS-2 intervention, and the abundance of short-chain fatty acid (SCFA)-producing bacteria (g__Ruminococcus_torques_group and g__Butyricicoccus) and bacteria with UA degradation capacity (g__unclassified_f__Lachnospiraceae and g__Negativibacillus) increased significantly; intestinal SCFAs, especially propionic acid, increased accordingly. These experimental data suggest that the beneficial effects of JS-2 may derive from changes in the gut microbiome, altering host-microbiota interactions, reducing UA levels by increasing UA excretion, and reducing absorption. These findings provided new evidence that JS-2 has the potential to be used as a naturally functional food for the prevention of HUA.
Collapse
Affiliation(s)
- Jiahui Wu
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Xiang Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Lvbu Aga
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Leimengyuan Tang
- Bayingolin Mongolian Autonomous Prefecture Institute for Food and Drug Control, No. 101, North Jianguo Road, Korla City 841000, Bayingolin Mongol Autonomous Prefecture, Xinjiang Uygur Autonomous Region, China
| | - Shuting Tan
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Dachuan Zhang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Houxier Li
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Li Yang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Nan Zhang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Shiyao Su
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Maochun Xiao
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Rongting Min
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Aji Li
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| | - Xueyong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China
| |
Collapse
|
14
|
Xue M, Du R, Zhou Y, Liu Y, Tian Y, Xu Y, Yan J, Song P, Wan L, Xu H, Zhang H, Liang H. Fucoidan Supplementation Relieved Kidney Injury and Modulated Intestinal Homeostasis in Hyperuricemia Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27187-27202. [PMID: 39600107 DOI: 10.1021/acs.jafc.4c07209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Hyperuricemia is a metabolic disease characterized by an excessively increased level of uric acid (UA) in the blood, with an increasing prevalence and often associated with kidney damage. Gut microbiota and endotoxins of gut origin are key mediators in the gut-kidney axis that can cause renal impairment. The study was to reveal the protective effects of fucoidan on renal injury caused by hyperuricemia. The hyperuricemia model was established in C57BL/6J mice. After 10 weeks of fucoidan supplementation, we found that the levels of serum UA and creatinine were reduced, and the levels of renal tumor necrosis factor α, interleukin-18 (IL-18), IL-6, and interleukin-1β (IL-1β) were also decreased. Fucoidan inhibited the expressions of phosphorylated NF-κB p65, NLRP3, and activated caspase-1 in the kidneys. Fucoidan also regulated the expressions of Bcl-2 family proteins and decreased the activation of caspase-3, thereby exerting antiapoptotic effect. In addition, fucoidan could reduce the expressions of glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1) proteins, thereby promoting the excretion of UA from the kidneys. Moreover, the protective effect of fucoidan on renal injury may be related to maintaining intestinal homeostasis. Fucoidan reduced serum lipopolysaccharide and improved the intestinal mucosal barrier function. Fucoidan decreased the abundances of Blautia, Muribaculaceae, and Dubosiella, and increased the abundances of Lactobacillus. High-dose fucoidan supplementation increased the content of butyric acid and enhanced the expression of ATP binding box transporter G2 (ABCG2) via the AMPK/AKT/CREB pathway in ileum. Conclusion: Fucoidan could protect against hyperuricemia-induced renal injury by inhibiting renal inflammation and apoptosis and modulating intestinal homeostasis in hyperuricemia mice.
Collapse
Affiliation(s)
- Meilan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Ronghuan Du
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Yifan Zhou
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, P. R. China
| | - Yuhan Liu
- School of Biomedical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yingjie Tian
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Yan Xu
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Jiayi Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Pengzhao Song
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Lu Wan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Hongsen Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, P. R. China
| | - Huaqi Zhang
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| | - Hui Liang
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao 266071, P. R. China
| |
Collapse
|
15
|
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
|
16
|
Bi C, Zhang L, Liu J, Chen L. Lactobacillus paracasei 259 alleviates hyperuricemia in rats by decreasing uric acid and modulating the gut microbiota. Front Nutr 2024; 11:1450284. [PMID: 39600720 PMCID: PMC11588492 DOI: 10.3389/fnut.2024.1450284] [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: 06/17/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Hyperuricemia (HUA) is a metabolic disease arising from abnormal purine metabolism. It contributes to an increased risk of kidney damage. The present study aimed to investigate the uric acid (UA)-lowering effects of Lactobacillus paracasei 259 isolated from yak yogurt and explore its underlying mechanisms. Our results revealed that L. paracasei 259 decreased the UA levels in rats and inhibited the serum activities of xanthine oxidase. In addition, L. paracasei 259 reduced the levels of pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6) in the kidney and altered the expressions of UA transporters (ABC transporter 2 (ABCG2), PDZ domain containing 1 (PDZK1), urate transporter 1 (URAT1), and sodium-phosphate cotransporter type 4 (NPT4)) to near normal levels. Moreover, it increased the abundance of beneficial bacteria in the gut and recovered the gut microbiota composition, promoting the production of short-chain fatty acids (SCFAs). These findings suggested that L. paracasei 259 can potentially be used to decrease UA levels, repair kidney damage, regulate gut microbiota, and alleviate HUA.
Collapse
Affiliation(s)
| | | | | | - Lianhong Chen
- College of Food Science and Technology, Southwest Minzu University, Chengdu, China
| |
Collapse
|
17
|
Fu Y, Luo XD, Li JZ, Mo QY, Wang X, Zhao Y, Zhang YM, Luo HT, Xia DY, Ma WQ, Chen JY, Wang LH, Deng QY, Ben L, Kashif Saleemi M, Jiang XZ, Chen J, Miao K, Lin ZP, Zhang P, Ye H, Cao QY, Zhu YW, Yang L, Tu Q, Wang W. Host-derived Lactobacillus plantarum alleviates hyperuricemia by improving gut microbial community and hydrolase-mediated degradation of purine nucleosides. eLife 2024; 13:e100068. [PMID: 39508089 PMCID: PMC11542919 DOI: 10.7554/elife.100068] [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: 06/13/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
The gut microbiota is implicated in the pathogenesis of hyperuricemia (HUA) and gout. However, it remains unclear whether probiotics residing in the host gut, such as Lactobacillus, can prevent HUA development. Herein, we isolated Lactobacillus plantarum SQ001 from the cecum of HUA geese and conducted in vitro assays on uric acid (UA) and nucleoside co-culture. Metabolomics and genome-wide analyses, revealed that this strain may promote nucleoside uptake and hydrolysis through its nucleoside hydrolase gene. The functional role of iunH gene was confirmed via heterologous expression and gene knockout studies. Oral administration of L. plantarum SQ001 resulted in increased abundance of Lactobacillus species and reduced serum UA levels. Furthermore, it downregulated hepatic xanthine oxidase, a key enzyme involved in UA synthesis, as well as renal reabsorption protein GLUT9, while enhancing the expression of renal excretion protein ABCG2. Our findings suggest that L. plantarum has potential to ameliorate gut microbial dysbiosis with HUA, thereby offering insights into its potential application as a probiotic therapy for individuals with HUA or gout.
Collapse
Affiliation(s)
- Yang Fu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Xiao-Dan Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Jin-Ze Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Qian-Yuan Mo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Xue Wang
- State Key Laboratory of Microbial Technology, Shandong UniversityShandongChina
| | - Yue Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - You-Ming Zhang
- State Key Laboratory of Microbial Technology, Shandong UniversityShandongChina
| | - Hao-Tong Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Dai-Yang Xia
- School of Marine Sciences, Sun Yat-sen University, and Southern Marine Science and Engineering Guangdong LaboratoryZhuhaiChina
| | - Wei-Qing Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Jian-Ying Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Li-Hau Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Qiu-Yi Deng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Lukuyu Ben
- International Livestock Research InstituteNairobiKenya
| | | | - Xian-Zhi Jiang
- Microbiome Research Center, Moon (Guangzhou) Biotech Co. LtdGuangdongChina
| | - Juan Chen
- Microbiome Research Center, Moon (Guangzhou) Biotech Co. LtdGuangdongChina
| | - Kai Miao
- CancerCenter, Faculty of Health Sciences, University of MacauMacauChina
| | - Zhen-Ping Lin
- Shantou Baisha Research Institute of Origin Species of Poultry and StockShantouChina
| | - Peng Zhang
- Chimelong Safari Park, Chimelong Group CoGuangzhouChina
| | - Hui Ye
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Qing-Yun Cao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Yong-Wen Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Lin Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| | - Qiang Tu
- Helmholtz International Lab for Anti-Infectives, Shandong University–Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, Shandong UniversityQingdaoChina
- Shenzhen Key Laboratory of Genome Manipulation and Biosynthesis, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Wence Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural UniversityGuangzhouChina
| |
Collapse
|
18
|
Wu L, Chen J, Yu Q, Lu C, Shu Y. Hypoxanthine Produces Rapid Antidepressant Effects by Suppressing Inflammation in Serum and Hippocampus. ACS Chem Neurosci 2024; 15:3970-3980. [PMID: 39441118 DOI: 10.1021/acschemneuro.4c00345] [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: 10/25/2024] Open
Abstract
The occurrence and development of depression are closely related to disorders of the brain and peripheral substances. Abnormal metabolites in the blood affect the signal regulation function of the nerve center, which is one of the key factors for depression episodes. This study was focused on metabolites in serum and the mechanism of its antidepressant in the hippocampus. In the present study, serum metabolites in patients with depression were screened by metabolomic techniques. Various depressive mouse models and behavioral tests were used to assess its antidepressant effects. The expressions of inflammatory signaling were detected by using Western blot, ELISA, and immunofluorescence. We found that the metabolite hypoxanthine in the serum of patients with depression was significantly reduced, and the same result was also found in two mouse models of depression such as chronic unpredictable mild stress (CUMS) and social defeat stress (SD). By administering different doses of hypoxanthine (5, 10, 15 mg/kg), we found that only 15 mg/kg was able to significantly reduce the latency and increase food consumption in the novelty suppressed-feeding test (NSF), which was also able to reverse the depressive phenotypes of mice in the CUMS model after a single administration at 2 h later. Hypoxanthine obviously reduced the expressions of inflammation in serum and downregulated the expressions of MAPK and NLRP3-related pathways in the hippocampus in CUMS mice. Moreover, hypoxanthine also suppressed the activations of glial cells including GFAP and IBA-1 in hippocampal CA1, CA3, and dentate gyrus (DG). To sum up, hypoxanthine exerted antidepressant effect relying on the inhibition of peripheral and hippocampal inflammations by regulating MAPK, NLRP3-related pathways, and glial cells. This was the first time that we have found a disordered metabolite in patients with depression and further systematically demonstrated its efficacy and potential mechanism of antidepressants, providing new ideas for antidepressant drug development.
Collapse
Affiliation(s)
- Lei Wu
- Department of Pharmacy, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Avenue, Qinhuai District, Nanjing 210029, P. R. China
| | - Jianhuai Chen
- Department of Pharmacy, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Avenue, Qinhuai District, Nanjing 210029, P. R. China
| | - Qiao Yu
- Department of Reproductive Center, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huaian 223300, P. R. China
| | - Chao Lu
- Department of Pharmacy, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Avenue, Qinhuai District, Nanjing 210029, P. R. China
| | - Yachun Shu
- Department of Pharmacy, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, 155 Hanzhong Avenue, Qinhuai District, Nanjing 210029, P. R. China
| |
Collapse
|
19
|
Wei H, Zhan L, Lv X, Lin Y, Zheng J, Yang W, Liu J, Sun J, Chen S. Gut commensal Parabacteroides distasonis exerts neuroprotective effects in acute ischemic stroke with hyperuricemia via regulating gut microbiota-gut-brain axis. J Transl Med 2024; 22:999. [PMID: 39501312 PMCID: PMC11539330 DOI: 10.1186/s12967-024-05800-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Hyperuricemia is considered as an independent risk factor for acute ischemic stroke (AIS), and some AIS patients are accompanied by an increase in serum uric acid. Recent studies have highlighted the important role of gut microbiota in both hyperuricemia and AIS, but there is little available data on the relationship between gut microbiota and the pathogenesis of AIS with hyperuricemia (HAS). METHODS Here we profiled the gut microbiota composition in 63 HAS patients and 269 non-HAS patients through 16s rRNA sequencing. Male rat with hyperuricemia were subjected to middle cerebral artery occlusion (MCAO) to establish HAS model and were then treated with Parabacteroides distasonis. Subsequently, the neurological deficit, pathological damages and blood-brain barrier disruption were evaluated. Moreover, the levels of ROS, inflammatory cytokines, NF-𝜿B pathway related protein, and vascular density markers were determined. RESULTS There were significant differences of gut microbiota composition between HAS patients and non-HAS patients, and a significant decrease in the abundance of Parabacteroides in HAS patients compared to non-HAS patients. Animal experiments showed that supplementation with P. distasonis increased beneficial commensal bacteria, significantly improved neurological deficits, pathological damages and BBB disruption, as well as reduced the level of serum uric acid in HAS rats. We further demonstrated that P. distasonis treatment decreased ROS level and increased SOD2 level, thereby reducing oxidative stress. Meanwhile, P. distasonis effectively inhibited NF-𝜿B signal pathway and reduced the production of inflammatory cytokines, including TNF-α and IL-1β, alleviating the inflammatory response. Notably, P. distasonis treatment increased the levels of vascular density markers including cluster of differentiation 31 (CD31) and alpha-smooth muscle actin (α-SMA), ameliorating vascular damage in HAS rats. CONCLUSIONS Together, these findings highlighted the important role of P. distasonis in the pathogenesis of HAS, and its mechanism was involved in the regulation of gut microbiota-gut-brain axis, which implied a novel strategy against HAS.
Collapse
Affiliation(s)
- Hongming Wei
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Lu Zhan
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xinhuang Lv
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yan Lin
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jie Zheng
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenwen Yang
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jing Sun
- Department of Geriatrics, the Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Songfang Chen
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
20
|
Chen YJ, Guo ZT, Chen HQ, Zhang SF, Bao YX, Xie Z, Ke JL, Ye WJ, Liang JC, Chen JC, Li N, Zheng FX, Liao H, Wu T, Pang JX. Salinomycin, a potent inhibitor of XOD and URAT1, ameliorates hyperuricemic nephropathy by activating NRF2, modulating the gut microbiota, and promoting SCFA production. Chem Biol Interact 2024; 403:111220. [PMID: 39222901 DOI: 10.1016/j.cbi.2024.111220] [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: 04/10/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Long-term hyperuricemia can induce kidney damage, clinically referred to as hyperuricemic nephropathy (HN), which is characterized by renal fibrosis, inflammation, and oxidative stress. However, currently used uric acid-lowering drugs are not capable of protecting the kidneys from damage. Therefore, uric acid-lowering drugs that can also protect the kidneys are urgently needed. In this study, we first discovered that salinomycin, an antibiotic, can regulate uric acid homeostasis and ameliorate kidney damage in mice with HN. Mechanistically, salinomycin inhibited serum and hepatic xanthine oxidase (XOD) activities and downregulated renal urate transporter 1 (URAT1) expression and transport activity, thus exerting uric acid-lowering effects in mice with HN. Furthermore, we found that salinomycin promoted p-NRF2 Ser40 expression, resulting in increased nuclear translocation of NRF2 and activation of NRF2. More importantly, salinomycin affected the gut microbiota and promoted the generation of short-chain fatty acids (SCFAs) in mice with HN. In conclusion, our results revealed that salinomycin maintains uric acid homeostasis and alleviates kidney injury in mice with HN by multiple mechanisms, suggesting that salinomycin might be a desirable candidate for HN treatment in the clinic.
Collapse
Affiliation(s)
- Yong-Jun Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zi-Tao Guo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hai-Qiao Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shi-Fan Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying-Xia Bao
- Baiyunshan Pharmaceutical General Factory, Guangzhou Baiyunshan Pharmaceutical Holdings Co., Ltd., Guangzhou, 510515, China; Key Laboratory of Key Technology Research on Chemical Raw Materials and Preparations of Guangdong Province, Guangzhou, 510515, China
| | - Zhoufan Xie
- Baiyunshan Pharmaceutical General Factory, Guangzhou Baiyunshan Pharmaceutical Holdings Co., Ltd., Guangzhou, 510515, China; Key Laboratory of Key Technology Research on Chemical Raw Materials and Preparations of Guangdong Province, Guangzhou, 510515, China
| | - Jia-le Ke
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Jie Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Cheng Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Chen Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ning Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Feng-Xin Zheng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hui Liao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ting Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Jian-Xin Pang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
21
|
Zhou F, Liu Y, Shi Y, Wu N, Xie Y, Zhou X. Association between gut microbiota and acute pancreatitis: a bidirectional Mendelian randomization study. J Gastroenterol Hepatol 2024; 39:1895-1902. [PMID: 38888069 DOI: 10.1111/jgh.16658] [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] [Received: 01/17/2024] [Revised: 04/07/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIM The dysbiosis of gut microbiota has been reported in acute pancreatitis. However, the direction and magnitude between host microbiota and pancreas remains to be established. This study investigated the association between gut microbiota and acute pancreatitis using Mendelian randomization (MR) methods. METHODS Summary statistics of gut microbiota abundance and acute pancreatitis were extracted from genome-wide association studies (GWAS). The two-sample bidirectional MR design was employed to assess genetic association between the microbiota and pancreatitis, followed by a comprehensive sensitivity analysis to verify the robustness of the results. RESULTS Seven microbiota taxa have been identified as significantly associated with the development of pancreatitis. Host genetic-driven order Bacteroidales and class Bacteroidia are associated with an increased risk of pancreatitis. The genera Coprococcus and Eubacterium fissicatena group also exhibit a positive effect on the development of pancreatitis, while the genera Prevotella, Ruminiclostridium, and Ruminococcaceae act as protective factors against pancreatitis. In contrast, acute pancreatitis was positively correlated with phylum Proteobacteria and genus Lachnospiraceae and negatively correlated with genus Holdemania. CONCLUSIONS The bidirectional relationship between gut microbiota and acute pancreatitis suggests a critical role for host-microbiota crosstalk in the development of the disease. Targeted modulation of specific gut microbiota enables the prevention and treatment of acute pancreatitis.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yang Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yanqing Shi
- Department of Gastroenterology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
| | - Nanzhen Wu
- Department of Gastrointestinal Surgery, Fengcheng People's Hospital, Fengcheng, Jiangxi Province, China
| | - Yong Xie
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaojiang Zhou
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
22
|
Liu X, Liang XQ, Lu TC, Feng Z, Zhang M, Liao NQ, Zhang FL, Wang B, Wang LS. Leech Poecilobdella manillensis protein extract ameliorated hyperuricemia by restoring gut microbiota dysregulation and affecting serum metabolites. World J Gastroenterol 2024; 30:3488-3510. [PMID: 39156502 PMCID: PMC11326090 DOI: 10.3748/wjg.v30.i29.3488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/20/2024] [Accepted: 07/19/2024] [Indexed: 07/29/2024] Open
Abstract
BACKGROUND Hyperuricemia (HUA) is a public health concern that needs to be solved urgently. The lyophilized powder of Poecilobdella manillensis has been shown to significantly alleviate HUA; however, its underlying metabolic regulation remains unclear. AIM To explore the underlying mechanisms of Poecilobdella manillensis in HUA based on modulation of the gut microbiota and host metabolism. METHODS A mouse model of rapid HUA was established using a high-purine diet and potassium oxonate injections. The mice received oral drugs or saline. Additionally, 16S rRNA sequencing and ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry-based untargeted metabolomics were performed to identify changes in the microbiome and host metabolome, respectively. The levels of uric acid transporters and epithelial tight junction proteins in the renal and intestinal tissues were analyzed using an enzyme-linked immunosorbent assay. RESULTS The protein extract of Poecilobdella manillensis lyophilized powder (49 mg/kg) showed an enhanced anti-trioxypurine ability than that of allopurinol (5 mg/kg) (P < 0.05). A total of nine bacterial genera were identified to be closely related to the anti-trioxypurine activity of Poecilobdella manillensis powder, which included the genera of Prevotella, Delftia, Dialister, Akkermansia, Lactococcus, Escherichia_Shigella, Enterococcus, and Bacteroides. Furthermore, 22 metabolites in the serum were found to be closely related to the anti-trioxypurine activity of Poecilobdella manillensis powder, which correlated to the Kyoto Encyclopedia of Genes and Genomes pathways of cysteine and methionine metabolism, sphingolipid metabolism, galactose metabolism, and phenylalanine, tyrosine, and tryptophan biosynthesis. Correlation analysis found that changes in the gut microbiota were significantly related to these metabolites. CONCLUSION The proteins in Poecilobdella manillensis powder were effective for HUA. Mechanistically, they are associated with improvements in gut microbiota dysbiosis and the regulation of sphingolipid and galactose metabolism.
Collapse
Affiliation(s)
- Xia Liu
- Medical College, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
- Department of Traditional Chinese Medicine, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People’s Hospital of Nanning, Nanning 530023, Guangxi Zhuang Autonomous Region, China
| | - Xing-Qiu Liang
- Department of Science and Technology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, Guangxi Zhuang Autonomous Region, China
| | - Tian-Cai Lu
- General Manager’s Office, Guangxi Fuxinyi Biological Technology Co. Ltd., Pingnan 537300, Guangxi Zhuang Autonomous Region, China
| | - Zhe Feng
- Department of Joint and Sports Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning 530011, Guangxi Zhuang Autonomous Region, China
| | - Min Zhang
- Department of Gerontology, Nanning Social Welfare Hospital, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Nan-Qing Liao
- Medical College, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Feng-Lian Zhang
- Medical College, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Bo Wang
- Medical College, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Li-Sheng Wang
- Medical College, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
23
|
Fu H, Chen Z, Teng W, Du Z, Zhang Y, Ye X, Yu Z, Zhang Y, Pi X. Effects of fructooligosaccharides and Saccharomyces boulardii on the compositional structure and metabolism of gut microbiota in students. Microbiol Res 2024; 285:127741. [PMID: 38761487 DOI: 10.1016/j.micres.2024.127741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 05/20/2024]
Abstract
Fructooligosaccharides (FOS) are a common prebiotic widely used in functional foods. Meanwhile, Saccharomyces boulardii is a fungal probiotic frequenly used in the clinical treatment of diarrhea. Compared with single use, the combination of prebiotics and probiotics as symbiotics may be more effective in regulating gut microbiota as recently reported in the literature. The present study aimed to investigate the effects of FOS, S. boulardii and their combination on the structure and metabolism of the gut microbiota in healthy primary and secondary school students using an in vitro fermentation model. The results indicated that S. boulardii alone could not effectively regulate the community structure and metabolism of the microbiota. However, both FOS and the combination of FOS and S. boulardii could effectively regulate the microbiota, significantly inhibiting the growth of Escherichia-Shigella and Bacteroides, and controlling the production of the gases including H2S and NH3. In addition, both FOS and the combination could significantly promote the growth of Bifidobacteria and Lactobacillus, lower environmental pH, and enhance several physiological functions related to synthesis and metabolism. Nevertheless, the combination had more unique benefits as it promoted the growth of Lactobacillus, significantly increased CO2 production and enhanced the functional pathways of carbon metabolism and pyruvic acid metabolism. These findings provide guidance for clinical application and a theoretical basis for the development of synbiotic preparations.
Collapse
Affiliation(s)
- Hao Fu
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China
| | - Zhixian Chen
- National Key Laboratory of Agricultural Microbiology, Angel Yeast Co., Ltd., Yichang 443003, PR China; The Hubei Provincial Key Laboratory of Yeast Function, Angel Yeast Co., Ltd., Yichang 443003, PR China; Yi Chang Engineering and Technology Research Center of Nutrition and Health Food, Angel Yeast Co., Ltd., Yichang 443003, PR China
| | - Weilin Teng
- Department of infectious Disease Control and Prevention, HangZhou Center for Disease Control and Prevention, Hangzhou 310006, PR China
| | - Zhi Du
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, PR China
| | - Yan Zhang
- National Key Laboratory of Agricultural Microbiology, Angel Yeast Co., Ltd., Yichang 443003, PR China; The Hubei Provincial Key Laboratory of Yeast Function, Angel Yeast Co., Ltd., Yichang 443003, PR China; Yi Chang Engineering and Technology Research Center of Nutrition and Health Food, Angel Yeast Co., Ltd., Yichang 443003, PR China
| | - Xiaoli Ye
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, PR China
| | - Zaichun Yu
- College of Bioengineering, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Yinjun Zhang
- College of Bioengineering, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Xionge Pi
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China; Institute of Rural Development, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China.
| |
Collapse
|
24
|
Han C, He C, Ding X, Li Z, Peng T, Zhang C, Chen H, Zuo Z, Huang J, Hu W. WWC1 upregulation accelerates hyperuricemia by reduction in renal uric acid excretion through Hippo signaling pathway. J Biol Chem 2024; 300:107485. [PMID: 38906255 PMCID: PMC11301351 DOI: 10.1016/j.jbc.2024.107485] [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: 01/24/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/23/2024] Open
Abstract
Hyperuricemia (HUA) is a metabolic disorder characterized by elevated serum uric acid (UA), primarily attributed to the hepatic overproduction and renal underexcretion of UA. Despite the elucidation of molecular pathways associated with this underexcretion, the etiology of HUA remains largely unknown. In our study, using by Uox knockout rats, HUA mouse, and cell line models, we discovered that the increased WWC1 levels were associated with decreased renal UA excretion. Additionally, using knockdown and overexpression approaches, we found that WWC1 inhibited UA excretion in renal tubular epithelial cells. Mechanistically, WWC1 activated the Hippo pathway, leading to phosphorylation and subsequent degradation of the downstream transcription factor YAP1, thereby impairing the ABCG2 and OAT3 expression through transcriptional regulation. Consequently, this reduction led to a decrease in UA excretion in renal tubular epithelial cells. In conclusion, our study has elucidated the role of upregulated WWC1 in renal tubular epithelial cells inhibiting the excretion of UA in the kidneys and causing HUA.
Collapse
Affiliation(s)
- Changshun Han
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chengyong He
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyan Ding
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zixuan Li
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Tianyun Peng
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chensong Zhang
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Haibing Chen
- Department of Endocrinology and Metabolism, Shanghai 10th People's Hospital, Tongji University, Shanghai, China
| | - Zhenghong Zuo
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiyi Huang
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Weiping Hu
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
25
|
Liu W, Nan F, Liu F, Yang X, Li Z, Jiang S, Zhang X, Li J, Yu M, Wang Y, Wang B. Isolation and identification of uric acid-dependent Aciduricibacillus chroicocephali gen. nov., sp. nov. from seagull feces and implications for hyperuricemia treatment. mSphere 2024; 9:e0002524. [PMID: 38814072 PMCID: PMC11332149 DOI: 10.1128/msphere.00025-24] [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: 01/10/2024] [Accepted: 04/10/2024] [Indexed: 05/31/2024] Open
Abstract
Hyperuricemia has become the second most prevalent metabolic disease after diabetes, but the limitations of urate-lowering treatment (ULT) drugs and patient nonadherence make ULT far less successful. Thus, more ULT approaches urgently need to be explored. Uric acid-degrading bacteria have potential application value in ULT. In this study, we isolated 44XBT, a uric acid-degrading bacterium, from black-headed gull (Chroicocephalus ridibundus) feces. Using a polyphasic taxonomic approach, strain 44XBT was identified as a novel genus within the family Bacillaceae; subsequently, the name Aciduricibacillus chroicocephali was proposed. Strain 44XBT had a unique uric acid-dependent phenotype and utilized uric acid and allantoin as the sole carbon and nitrogen sources, but not common carbon sources or complex media. In the genome, multiple copies of genes involved in uric acid metabolic pathway (pucL, pucM, uraD, and allB) were found. Six copies of pucL (encoding urate oxidase) were detected. Of these, five pucL copies were in a tandem arrangement and shared 70.42%-99.70% amino acid identity. In vivo experiments revealed that 44XBT reduced serum uric acid levels and attenuated kidney damage in hyperuricemic mice through uric acid catalysis in the gut and gut microbiota remodeling. In conclusion, our findings discover a strain for studying bacterial uric acid metabolism and may provide valuable insights into ULT. IMPORTANCE The increasing disease burden of hyperuricemia highlights the need for new therapeutic drugs and treatment strategies. Our study describes the developmental and application values of natural uric acid-degrading bacteria found in the gut of birds and broadened the source of bacteria with potential therapeutic value. Furthermore, the special physiology characteristics and genomic features of strain 44XBT are valuable for further study.
Collapse
Affiliation(s)
- Wenxuan Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fulong Nan
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fengjun Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaoli Yang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zonghui Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shasha Jiang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xianjuan Zhang
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Meng Yu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
26
|
Zhu W, Bi S, Fang Z, Iddrisu L, Deng Q, Sun L, Gooneratne R. Priestia megaterium ASC-1 Isolated from Pickled Cabbage Ameliorates Hyperuricemia by Degrading Uric Acid in Rats. Microorganisms 2024; 12:832. [PMID: 38674776 PMCID: PMC11052324 DOI: 10.3390/microorganisms12040832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Pickled cabbage, a traditional fermented food rich in functional microorganisms, can effectively control hyperuricemia and gout. In this study, a Priestia megaterium ASC-1 strain with strong uric acid (UA) degradation ability was isolated from pickled cabbage. After oral administration for 15 days, ASC-1 was stably colonized in the rats in this study. ASC-1 significantly reduced UA levels (67.24%) in hyperuricemic rats. Additionally, ASC-1 alleviated hyperuricemia-related inflammatory response, oxidative stress, and blood urea nitrogen. Intestinal microbial diversity results showed that ASC-1 restored intestinal injury and gut flora dysbiosis caused by hyperuricemia. These findings suggest that P. megaterium ASC-1 may be used as a therapeutic adjuvant for the treatment of hyperuricemia.
Collapse
Affiliation(s)
- Wenjuan Zhu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology, Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (W.Z.); (S.B.); (L.I.); (Q.D.); (L.S.)
| | - Siyuan Bi
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology, Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (W.Z.); (S.B.); (L.I.); (Q.D.); (L.S.)
| | - Zhijia Fang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology, Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (W.Z.); (S.B.); (L.I.); (Q.D.); (L.S.)
| | - Lukman Iddrisu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology, Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (W.Z.); (S.B.); (L.I.); (Q.D.); (L.S.)
| | - Qi Deng
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology, Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (W.Z.); (S.B.); (L.I.); (Q.D.); (L.S.)
| | - Lijun Sun
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology, Research Center of Marine Food, Key Laboratory of Advanced Processing of Aquatic Products of Guangdong Higher Education Institution, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (W.Z.); (S.B.); (L.I.); (Q.D.); (L.S.)
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Lincoln University, Lincoln 7647, Canterbury, New Zealand;
| |
Collapse
|
27
|
Zhou Y, Zeng Y, Wang R, Pang J, Wang X, Pan Z, Jin Y, Chen Y, Yang Y, Ling W. Resveratrol Improves Hyperuricemia and Ameliorates Renal Injury by Modulating the Gut Microbiota. Nutrients 2024; 16:1086. [PMID: 38613119 PMCID: PMC11013445 DOI: 10.3390/nu16071086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Resveratrol (RES) has been reported to prevent hyperuricemia (HUA); however, its effect on intestinal uric acid metabolism remains unclear. This study evaluated the impact of RES on intestinal uric acid metabolism in mice with HUA induced by a high-fat diet (HFD). Moreover, we revealed the underlying mechanism through metagenomics, fecal microbiota transplantation (FMT), and 16S ribosomal RNA analysis. We demonstrated that RES reduced the serum uric acid, creatinine, urea nitrogen, and urinary protein levels, and improved the glomerular atrophy, unclear renal tubule structure, fibrosis, and renal inflammation. The results also showed that RES increased intestinal uric acid degradation. RES significantly changed the intestinal flora composition of HFD-fed mice by enriching the beneficial bacteria that degrade uric acid, reducing harmful bacteria that promote inflammation, and improving microbial function via the upregulation of purine metabolism. The FMT results further showed that the intestinal microbiota is essential for the effect of RES on HUA, and that Lactobacillus may play a key role in this process. The present study demonstrated that RES alleviates HFD-induced HUA and renal injury by regulating the gut microbiota composition and the metabolism of uric acid.
Collapse
Affiliation(s)
- Yuqing Zhou
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yupeng Zeng
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Ruijie Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Juan Pang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Xin Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Zhijun Pan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yufeng Jin
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yu Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yan Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| |
Collapse
|
28
|
Zhao X, Cai P, Xiong S, Wei B, Du T, Huang T, Yu Q, Xie M, Xiong T. Lacticaseibacillus rhamnosus NCUH061012 alleviates hyperuricemia via modulating gut microbiota and intestinal metabolites in mice. FOOD BIOSCI 2024; 58:103699. [DOI: 10.1016/j.fbio.2024.103699] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
29
|
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
|
30
|
Fu Y, Chen YS, Xia DY, Luo XD, Luo HT, Pan J, Ma WQ, Li JZ, Mo QY, Tu Q, Li MM, Zhao Y, Li Y, Huang YT, Chen ZX, Li ZJ, Bernard L, Dione M, Zhang YM, Miao K, Chen JY, Zhu SS, Ren J, Zhou LJ, Jiang XZ, Chen J, Lin ZP, Chen JP, Ye H, Cao QY, Zhu YW, Yang L, Wang X, Wang WC. Lactobacillus rhamnosus GG ameliorates hyperuricemia in a novel model. NPJ Biofilms Microbiomes 2024; 10:25. [PMID: 38509085 PMCID: PMC10954633 DOI: 10.1038/s41522-024-00486-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/06/2024] [Indexed: 03/22/2024] Open
Abstract
Hyperuricemia (HUA) is a metabolic syndrome caused by abnormal purine metabolism. Although recent studies have noted a relationship between the gut microbiota and gout, whether the microbiota could ameliorate HUA-associated systemic purine metabolism remains unclear. In this study, we constructed a novel model of HUA in geese and investigated the mechanism by which Lactobacillus rhamnosus GG (LGG) could have beneficial effects on HUA. The administration of antibiotics and fecal microbiota transplantation (FMT) experiments were used in this HUA goose model. The effects of LGG and its metabolites on HUA were evaluated in vivo and in vitro. Heterogeneous expression and gene knockout of LGG revealed the mechanism of LGG. Multi-omics analysis revealed that the Lactobacillus genus is associated with changes in purine metabolism in HUA. This study showed that LGG and its metabolites could alleviate HUA through the gut-liver-kidney axis. Whole-genome analysis, heterogeneous expression, and gene knockout of LGG enzymes ABC-type multidrug transport system (ABCT), inosine-uridine nucleoside N-ribohydrolase (iunH), and xanthine permease (pbuX) demonstrated the function of nucleoside degradation in LGG. Multi-omics and a correlation analysis in HUA patients and this goose model revealed that a serum proline deficiency, as well as changes in Collinsella and Lactobacillus, may be associated with the occurrence of HUA. Our findings demonstrated the potential of a goose model of diet-induced HUA, and LGG and proline could be promising therapies for HUA.
Collapse
Affiliation(s)
- Yang Fu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yong-Song Chen
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Dai-Yang Xia
- School of Marine Sciences, Sun Yat-sen University, and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519082, China
| | - Xiao-Dan Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Hao-Tong Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jie Pan
- Hunan Shihua Biotech Co. Ltd., Changsha, 410000, China
| | - Wei-Qing Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jin-Ze Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qian-Yuan Mo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qiang Tu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, China
| | - Meng-Meng Li
- School of Agricultural Science and Engineering, Liaocheng University, Liaocheng, 252000, China
| | - Yue Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yi-Teng Huang
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zhi-Xian Chen
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Zhen-Jun Li
- Key Laboratory of Carcinogenesis and Translational Research, Departments of Lymphoma, Radiology and Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, 100080, China
| | - Lukuyu Bernard
- International Livestock Research Institute, Nairobi, 00100, Kenya
| | - Michel Dione
- International Livestock Research Institute, Nairobi, 00100, Kenya
| | - You-Ming Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, China
| | - Kai Miao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau, SAR, China
| | - Jian-Ying Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Shan-Shan Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jie Ren
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Ling-Juan Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xian-Zhi Jiang
- Microbiome Research Center, Moon (Guangzhou) Biotech Co. Ltd., Guangzhou, 510535, China
| | - Juan Chen
- Microbiome Research Center, Moon (Guangzhou) Biotech Co. Ltd., Guangzhou, 510535, China
| | - Zhen-Ping Lin
- Shantou Baisha Research Institute of Origin Species of Poultry and Stock, Shantou, 515041, China
| | - Jun-Peng Chen
- Shantou Baisha Research Institute of Origin Species of Poultry and Stock, Shantou, 515041, China
| | - Hui Ye
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qing-Yun Cao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yong-Wen Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Lin Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Xue Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, Shandong, China.
| | - Wen-Ce Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
31
|
Xu YX, Liu LD, Zhu JY, Zhu SS, Ye BQ, Yang JL, Huang JY, Huang ZH, You Y, Li WK, He JL, Xia M, Liu Y. Alistipes indistinctus-derived hippuric acid promotes intestinal urate excretion to alleviate hyperuricemia. Cell Host Microbe 2024; 32:366-381.e9. [PMID: 38412863 DOI: 10.1016/j.chom.2024.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/10/2024] [Accepted: 02/02/2024] [Indexed: 02/29/2024]
Abstract
Hyperuricemia induces inflammatory arthritis and accelerates the progression of renal and cardiovascular diseases. Gut microbiota has been linked to the development of hyperuricemia through unclear mechanisms. Here, we show that the abundance and centrality of Alistipes indistinctus are depleted in subjects with hyperuricemia. Integrative metagenomic and metabolomic analysis identified hippuric acid as the key microbial effector that mediates the uric-acid-lowering effect of A. indistinctus. Mechanistically, A. indistinctus-derived hippuric acid enhances the binding of peroxisome-proliferator-activated receptor γ (PPARγ) to the promoter of ATP-binding cassette subfamily G member 2 (ABCG2), which in turn boosts intestinal urate excretion. To facilitate this enhanced excretion, hippuric acid also promotes ABCG2 localization to the brush border membranes in a PDZ-domain-containing 1 (PDZK1)-dependent manner. These findings indicate that A. indistinctus and hippuric acid promote intestinal urate excretion and offer insights into microbiota-host crosstalk in the maintenance of uric acid homeostasis.
Collapse
Affiliation(s)
- Ying-Xi Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Lu-Di Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jiang-Yuan Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Shan-Shan Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Bing-Qi Ye
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jia-Lu Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jing-Yi Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Zhi-Hao Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Yi You
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Wen-Kang Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jia-Lin He
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Yan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China.
| |
Collapse
|
32
|
Lu M, Yin J, Xu T, Dai X, Liu T, Zhang Y, Wang S, Liu Y, Shi H, Zhang Y, Mo F, Sukhorukov V, Orekhov AN, Gao S, Wang L, Zhang D. Fuling-Zexie formula attenuates hyperuricemia-induced nephropathy and inhibits JAK2/STAT3 signaling and NLRP3 inflammasome activation in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117262. [PMID: 37788785 DOI: 10.1016/j.jep.2023.117262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/24/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuling-Zexie (FZ) formula, a traditional Chinese herbal prescription composed of Poria cocos (Schwan.) Wolf. (Poria), Pueraria lobate (Willd.) Howe. (Puerariae Lobatae Radix), Alisma orientale (Sam.) Julep. (Alismatis Rhizoma), and Atractylodes lancea (Thunb.) Dc. (Atractylodis Rhizoma), has been clinically used to ameliorate hyperuricemia (HUA) and its associated renal injury. AIM OF STUDY This study aims to explore the action and mechanism of FZ on renal inflammation and dysfunction caused by HUA. MATERIALS AND METHODS FZ was orally administered to rapid HUA mouse induced by potassium oxonate (PO) and hypoxanthine (HX) for 7 days. Serum levels of uric acid (UA), creatinine (CRE), blood urea nitrogen (BUN), xanthine oxidase (XOD), adenosine deaminase (ADA), alanine aminotransferase (ALT), aspartate aminotransferase (AST), urine levels of UA, CRE and urinary albumin were determined by biochemical assays. Serum levels of interleukin (IL)-1β and IL-6 were tested by ELISA. Hematoxylin-eosin and Masson staining were used to examine kidney and liver histopathological alterations. The expressions of renal glucose transporter 9 (GLUT9), ATP-binding cassette subfamily G member 2 (ABCG2), organic anion transporter 1 (OAT1), phospho-janus kinase 2 (p-JAK2), p-signal transducer and activator of transcription 3 (p-STAT3), suppression of cytokine signaling 3 (SOCS3), NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein (ASC), and cleaved-cysteinyl aspartate specific proteinase-1 (cleaved-Cas-1) were detected by western blots. The potential protein targets and pathways of FZ intervention on HUA were predicted by network pharmacology. The constituents in FZ aqueous extract were analyzed by UPLC-MS. RESULTS FZ reduced serum UA, CRE, BUN, and urinary albumin and increased urine UA, CRE levels in HUA mice. In addition, the treatment with FZ to HUA mice inhibited the elevated serum levels of XOD and ADA, and regulated renal urate transports including OAT1, GLUT9 and ABCG2. FZ also attenuated kidney inflammation and fibrosis and downregulated the expressions of IL-1β, p-JAK2, p-STAT3, SOCS3, IL-6, NLRP3, ASC, and cleaved-Cas-1. Thirteen compounds were identified in the FG, including L-phenylalanine, D-tryptophan, 3'-hydroxypuerarin, Puerarin, 3'-Methoxy Puerarin, Daidzin, Pueroside A, formononetin-8-C- [xylosyl (1→6)]-glucoside, Ononin, Alisol I 23-acetate, 16-oxo-alisol A, Alisol C and Alisol A. CONCLUSION FZ inhibits serum UA generation and promotes urine UA excretion as well as attenuates kidney inflammation and fibrosis in HUA mouse with nephropathy. The underlying mechanism of its action may be associated with suppression of the JAK2/STAT3 signaling pathway and NLRP3 inflammasome activation. This formula may offer a novel source for developing anti-HUA drugs.
Collapse
Affiliation(s)
- Meixi Lu
- 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.
| | - 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.
| | - 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.
| | - 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.
| | - Yanfei Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Fangfang Mo
- 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
|
33
|
Liu X, Han CH, Mao T, Wu J, Ke LY, Guo YJ, Han RS, Tian ZB. Commensal Enterococcus faecalis W5 ameliorates hyperuricemia and maintains the epithelial barrier in a hyperuricemia mouse model. J Dig Dis 2024; 25:44-60. [PMID: 38126957 DOI: 10.1111/1751-2980.13249] [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] [Received: 06/08/2023] [Revised: 11/03/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE The intestine is responsible for approximately one-third of uric acid (UA) excretion. The effect of commensal Enterococcus faecalis (E. faecalis), one of the most colonized bacteria in the gut, on UA excretion in the intestine remains to be investigated. The aim of this study was to evaluate the effect of commensal E. faecalis on UA metabolism and gut microbiota. METHODS The 16S rRNA gene sequencing was used to examine the species of Enterococcus in mouse fecal content. E. faecalis strain was isolated from mouse feces and identified to be E. faecalis W5. The hyperuricemia (HUA) animal model was established with yeast-rich forage and 250 mg·kg-1 ·day-1 potassium oxonate. Oral administration of E. faecalis W5 was given for 20 days, serving as the Efa group. RESULTS Disrupted intestinal barrier, activated proinflammatory response and low UA excretion in the intestine were found in HUA mice. After E. faecalis W5 treatment, the gut barrier was restored and serum UA level was decreased. Additionally, fecal and intestinal UA levels were elevated, intestinal urate transporter ABCG2 and purine metabolism were upregulated. Moreover, short-chain fatty acid levels were increased, and intestinal inflammation was ameliorated. CONCLUSIONS Commensal E. faecalis W5 ameliorated HUA through reversing the impaired gut barrier, promoting intestinal UA secretion by regulating ABCG2 expression, and decreasing intestinal UA synthesis by regulating purine metabolism. The results may provide the potential for developing treatments for HUA through the intestine.
Collapse
Affiliation(s)
- Xin Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chun Hua Han
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Tao Mao
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Jie Wu
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Le Yong Ke
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Ying Jie Guo
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Rong Shuang Han
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Zi Bin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
34
|
Jia X, Chen Q, Wu H, Liu H, Jing C, Gong A, Zhang Y. Exploring a novel therapeutic strategy: the interplay between gut microbiota and high-fat diet in the pathogenesis of metabolic disorders. Front Nutr 2023; 10:1291853. [PMID: 38192650 PMCID: PMC10773723 DOI: 10.3389/fnut.2023.1291853] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
In the past two decades, the rapid increase in the incidence of metabolic diseases, including obesity, diabetes, dyslipidemia, non-alcoholic fatty liver disease, hypertension, and hyperuricemia, has been attributed to high-fat diets (HFD) and decreased physical activity levels. Although the phenotypes and pathologies of these metabolic diseases vary, patients with these diseases exhibit disease-specific alterations in the composition and function of their gut microbiota. Studies in germ-free mice have shown that both HFD and gut microbiota can promote the development of metabolic diseases, and HFD can disrupt the balance of gut microbiota. Therefore, investigating the interaction between gut microbiota and HFD in the pathogenesis of metabolic diseases is crucial for identifying novel therapeutic strategies for these diseases. This review takes HFD as the starting point, providing a detailed analysis of the pivotal role of HFD in the development of metabolic disorders. It comprehensively elucidates the impact of HFD on the balance of intestinal microbiota, analyzes the mechanisms underlying gut microbiota dysbiosis leading to metabolic disruptions, and explores the associated genetic factors. Finally, the potential of targeting the gut microbiota as a means to address metabolic disturbances induced by HFD is discussed. In summary, this review offers theoretical support and proposes new research avenues for investigating the role of nutrition-related factors in the pathogenesis of metabolic disorders in the organism.
Collapse
Affiliation(s)
- Xiaokang Jia
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Qiliang Chen
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huiwen Wu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Hongbo Liu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Chunying Jing
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Aimin Gong
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Yuanyuan Zhang
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
35
|
Chen T, Li B, Zheng K, Liu Y, Zhang Z, Hu H, Qian G, Jiang J. Lactobacillus paracasei R3 Alleviates Tumor Progression in Mice with Colorectal Cancer. Curr Microbiol 2023; 81:38. [PMID: 38091085 DOI: 10.1007/s00284-023-03525-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 07/25/2023] [Indexed: 12/18/2023]
Abstract
Lactobacillus paracasei (L. paracasei), a common probiotic lactobacillus, has important functions in the food industry and human health. However, different strains of L. paracasei inevitably show differences in activity and colonization resistance, leading to differentiation in their functions, as well as their physical or chemical properties. The purpose of this study was to evaluate the characteristics of L. paracasei R3 (L.p R3) isolated from healthy human feces and determine whether the criteria for edible probiotics is met. The hemolysis type, biofilm-forming ability, antibiotic susceptibility, toxicity, and effective activity of L.p R3 were determined by establishing its probiotic activity traits in vitro and in vivo. The results showed that L.p R3 had a moderate biofilm formation ability, was sensitive to 11 antibiotics, was resistant to eight antibiotics, and was not hemolytic. The culture characteristics, morphology, and biochemical responses of the strain were consistent with the seed batch characteristics. In toxicity assays, L.p R3-fed mice showed no abnormalities in body weight, growth, or various organs. Additionally, L.p R3 was found to be effective in the prevention and treatment of colorectal cancer. In conclusion, our results revealed that L.p R3 has potential value as an edible probiotic without toxic side effects and alleviated the tumor progression of colorectal cancer in mice.
Collapse
Affiliation(s)
- Tao Chen
- Center of Human Microecology Engineering and Technology of Guangdong Province, Guangdong Longsee Biomedical Corporation, Guangzhou, 510535, Guangdong, China
| | - Baoxia Li
- Center of Human Microecology Engineering and Technology of Guangdong Province, Guangdong Longsee Biomedical Corporation, Guangzhou, 510535, Guangdong, China
| | - Kangdi Zheng
- Center of Human Microecology Engineering and Technology of Guangdong Province, Guangdong Longsee Biomedical Corporation, Guangzhou, 510535, Guangdong, China
| | - Yan Liu
- Center of Human Microecology Engineering and Technology of Guangdong Province, Guangdong Longsee Biomedical Corporation, Guangzhou, 510535, Guangdong, China.
| | - Zhao Zhang
- Center of Human Microecology Engineering and Technology of Guangdong Province, Guangdong Longsee Biomedical Corporation, Guangzhou, 510535, Guangdong, China
| | - Huimei Hu
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
| | - Guoqiang Qian
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
| | - Jianwei Jiang
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
36
|
Zhou X, Zhang B, Zhao X, Zhang P, Guo J, Zhuang Y, Wang S. Coffee Leaf Tea Extracts Improve Hyperuricemia Nephropathy and Its Associated Negative Effect in Gut Microbiota and Amino Acid Metabolism in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:17775-17787. [PMID: 37936369 DOI: 10.1021/acs.jafc.3c02797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Hyperuricemia nephropathy (HN) is a metabolic disease characterized by tubular damage, tubulointerstitial fibrosis, and uric acid kidney stones and has been demonstrated to be associated with hyperuricemia. Coffee leaf tea is drunk as a functional beverage. However, its prevention effects on HN remain to be explored. This study showed that coffee leaf tea extracts (TE) contain 19 polyphenols, with a total content of 550.15 ± 27.58 mg GAE/g. TE decreased serum uric acid levels via inhibiting XOD activities and modulating the expression of urate transporters (GLUT9, OAT3, and ABCG2) in HN rats. TE prevented HN-induced liver and kidney damage and attenuated renal fibrosis. Moreover, it upregulated the abundance of SCFA-producing bacteria (Phascolarctobacterium, Alloprevotella, and Butyricicoccus) in the gut and reversed the amino acid-related metabolism disorder caused by HN. TE also decreased the circulating LPS and d-lactate levels and increased the fecal SCFA levels. This study supported the preliminary and indicative effect of coffee leaf tea in the prevention of hyperuricemia and HN.
Collapse
Affiliation(s)
- Xiaofei Zhou
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Bowei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| | - Xiuli Zhao
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Pixian Zhang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jingting Guo
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yuan Zhuang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
| |
Collapse
|
37
|
Wang D, Zhang L, He D, Zhang Y, Bao J, Gao W, Cheng W, Zhu C, Jin H, Zhang W, Zhu H, Pan H. Systemic pharmacology reveal the mechanism by which the Qiangjin Zhuanggu Qufeng mixture inhibits LPS-induced pyroptosis of rat nucleus pulposus cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154998. [PMID: 37523835 DOI: 10.1016/j.phymed.2023.154998] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 07/02/2023] [Accepted: 07/25/2023] [Indexed: 08/02/2023]
Abstract
OBJECTIVE Low back pain (LBP) is a worldwide health issue primarily attributed to intervertebral disc degeneration (IVDD). Qiangjin Zhuang Qufeng mixture (QJZG), an approved hospital-based formula with years of clinical application, has demonstrated notable therapeutic effects in the treatment of LBP. Nevertheless, the underlying mechanism by which it alleviates LBP remains uncertain. METHODS The bioactive constituents of QJZG were initially identified using ultra-performance liquid chromatography quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS/MS). Subsequently, network pharmacology was employed to explore the core components and targets. In vivo and in vitro experiments were then conducted to validate the specific mechanism of action of QJZG based on the identified targets and pathways. Following that, ultra-high-performance liquid chromatography/mass spectrometry combined with 16S rRNA gene sequencing of blood and faecal samples was utilized to assess the impact of gut microbiota on faecal and serum metabolites subsequent to QJZG administration in intervertebral disc degeneration (IVDD) rats. RESULTS The principal constituents of QJZG were identified using UPLC-Q-TOF-MS/MS, revealing a substantial enrichment of flavonoids and triterpenes. Network pharmacology analysis indicated the potential inhibitory effects of QJZG on the NLRP3 inflammasome and downstream inflammatory factors. Furthermore, investigations demonstrated that intervertebral disc degeneration may be attributed to pyroptotic cell death within the nucleus pulposus. In vitro experiments were performed utilizing LPS to induce the inflammatory response in nucleus pulposus cells (NPC), and it was observed that QJZG-containing serum significantly suppressed key pyroptosis-related genes and downstream inflammatory factors. Additionally, in vivo experiments substantiated the capacity of QJZG to preserve disc height and ameliorate the progression of disc degeneration. Concurrently, oral pharmacotherapy in animal studies prominently involved the effects of Enterobacteriaceae and Clostridium, closely intertwined with lipid metabolism. CONCLUSIONS QJZG exhibited a delaying effect on IVDD by preserving the equilibrium between extracellular matrix (ECM) synthesis and degradation in NPCs. This effect was achieved through the suppression of NLRP3 inflammasome expression and the prevention of pyroptosis in NPCs.
Collapse
Affiliation(s)
- Dong Wang
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China; Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou 310021, China; Institute of Orthopaedics and Traumatology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Liangping Zhang
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Du He
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Yujun Zhang
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Jianhang Bao
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Wenshuo Gao
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Wei Cheng
- Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou 310021, China
| | - Chengyue Zhu
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China; Institute of Orthopaedics and Traumatology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Hongting Jin
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China; Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei Zhang
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Hang Zhu
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China
| | - Hao Pan
- Department of Orthopaedics, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China; Department of Orthopaedics, Hangzhou Dingqiao Hospital, Huanding Road NO 1630, Hangzhou 310021, China; Institute of Orthopaedics and Traumatology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Tiyuchang Road NO 453, Hangzhou 310007, China.
| |
Collapse
|
38
|
Zhang Y, Li Y, Li C, Zhao Y, Xu L, Ma S, Lin F, Xie Y, An J, Wang S. Paeonia × suffruticosa Andrews leaf extract and its main component apigenin 7-O-glucoside ameliorate hyperuricemia by inhibiting xanthine oxidase activity and regulating renal urate transporters. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154957. [PMID: 37478683 DOI: 10.1016/j.phymed.2023.154957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/20/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Hyperuricemia is an important pathological basis of gout and a distinct hazard factor for metabolic syndromes and cardiovascular and chronic renal disease, but lacks safe and effective treatments currently. Paeonia × suffruticosa Andrews leaf effectively reduced serum uric acid in gout patients; however, the material foundation and the mechanism remain unclear. PURPOSE To determine the primary active components and mechanism of P. suffruticosa leaf in hyperuricemic mice. METHODS The chemical constituents of P. suffruticosa leaf was identified using high-performance liquid chromatographic analysis. The anti-hyperuricemic activity of P. suffruticosa leaf extract (12.5, 25, 50, 100, and 200 mg/kg) and its components was evaluated in hyperuricemic mice induced by a high purine diet for 14 days. Then, the urate-lowering effects of apigenin 7-O-glucoside (0.09, 0.18, and 0.36 mg/kg) were assessed in another hyperuricemic mice model built by administrating potassium oxonate and adenine for 4 weeks. The inhibitory effect of apigenin 7-O-glucoside on uric acid production was elucidated by investigating xanthine oxidase activity in vitro and in serum and the liver and through molecular docking. Immunofluorescence and western blot analyses of the expression of renal urate transporter 1 (URAT1), glucose transporter 9 (GLUT9), organic anion transporters 1 (OAT1), and ATP-binding cassette G member 2 (ABCG2) proteins elucidated how apigenin 7-O-glucoside promoted uric acid excretion. RESULTS Six compounds were identified in P. suffruticosa leaf: gallic acid, methyl gallate, oxypaeoniflorin, paeoniflorin, galloylpaeoniflorin, and apigenin 7-O-glucoside. P. suffruticosa leaf extract significantly attenuated increased serum uric acid, creatinine, and xanthine oxidase activity in hyperuricemic mice. Apigenin 7-O-glucoside from P. suffruticosa leaf reduced uric acid, creatinine, and malondialdehyde serum levels, increased superoxide dismutase activity, and partially restored the spleen coefficient in hyperuricemic mice. Apigenin 7-O-glucoside inhibited xanthine oxidase activity in vitro and decreased serum and liver xanthine oxidase activity and liver xanthine oxidase protein expression in hyperuricemic mice. Molecular docking revealed that apigenin 7-O-glucoside bound to xanthine oxidase. Apigenin 7-O-glucoside facilitated uric acid excretion by modulating the renal urate transporters URAT1, GLUT9, OAT1, and ABCG2. Apigenin 7-O-glucoside protected against renal damage and oxidative stress caused by hyperuricemia by reducing serum creatinine, blood urea nitrogen, malondialdehyde, and renal reactive oxygen species levels; increasing serum and renal superoxide dismutase activity; restoring the renal coefficient; and reducing renal pathological injury. CONCLUSION Apigenin 7-O-glucoside is the main urate-lowering active component of P. suffruticosa leaf extract in the hyperuricemic mice. It suppressed liver xanthine oxidase activity to decrease uric acid synthesis and modulated renal urate transporters to stimulate uric acid excretion, alleviating kidney damage caused by hyperuricemia.
Collapse
Affiliation(s)
- Yan Zhang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yao Li
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, China
| | - Chang Li
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Yani Zhao
- Xi'an Encephalopathy Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710000, China
| | - Lu Xu
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Shanbo Ma
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Fen Lin
- Research and Development Department, Shaanxi Fengdan Zhengyuan Biotechnology Limited Company, Xi'an, Shaanxi 710076, China
| | - Yanhua Xie
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Junming An
- Department of Acupuncture, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710021, China.
| | - Siwang Wang
- The College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
39
|
Zhou Z, Feng D, Shi D, Gao P, Wang L, Wu Z. Untargeted and targeted metabolomics reveal bile acid profile changes in rats with ethylene glycol-induced calcium oxalate nephrolithiasis. Chem Biol Interact 2023; 381:110570. [PMID: 37244400 DOI: 10.1016/j.cbi.2023.110570] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Calcium oxalate (CaOx) nephrolithiasis is a prevalent disorder linked to metabolism. Examining metabolic alterations could potentially give an initial understanding of the origins of CaOx nephrolithiasis. This study aims to determine gut metabolic biomarkers differentiating CaOx nephrolithiasis utilizing untargeted and targeted metabolomics. CaOx nephrolithiasis model rats were built by 1% ethylene glycol administration. Histologic staining and renal function measurement revealed the presence of crystals in the lumen of the renal tubules, the renal injury and interstitial fibrosis in CaOx rats, demonstrating that the models of CaOx were established successfully. Hematoxylin & eosin (H&E) staining showed that CaOx group had inflammation and damage in the ileal tissue. Immunofluorescence and PCR results displayed that the tight junction proteins, ZO-1 and Occludin levels were decreased in the ileal tissues of the CaOx group. The untargeted metabolomic analysis revealed that 269 gut metabolites were differentially expressed between the CaOx group and the control group. Meanwhile, bile secretion, the main metabolic pathway in CaOx nephrolithiasis, was identified. Following, five significant bile acid metabolites were selected utilizing the targeted bile acid metabolomics, including Hyodeoxycholic acid (HDCA), Glycohyodeoxycholic acid (GHDCA), Nor-Deoxycholic Acid, omega-muricholic acid, and Taurolithocholic acid. Among these metabolites, HDCA and GHDCA presented the highest predictive accuracy with AUC = 1 to distinguish the CaOx group from the control group. As a result of network pharmacology, target genes of HDCA and GHDCA in CaOx nephrolithiasis were enriched in oxidative stress and apoptosis pathways. Conclusively, our study provides insight into bile acids metabolic changes related to CaOx nephrolithiasis. Although alterations in biochemical pathways indicate a complex pathology in CaOx rats, bile acid changes may serve as biomarkers of CaOx nephrolithiasis.
Collapse
Affiliation(s)
- Zijian Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
| | - Dexiang Feng
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, 215123, PR China
| | - Donghui Shi
- Department of Urology, Suzhou Wu Zhong People's Hospital, Suzhou, 215100, PR China
| | - Peng Gao
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China
| | - Lujia Wang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China.
| | - Zhong Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, PR China; Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
40
|
Zhao H, Chen X, Zhang L, Tang C, Meng F, Zhou L, Zhu P, Lu Z, Lu Y. Ingestion of Lacticaseibacillus rhamnosus Fmb14 prevents depression-like behavior and brain neural activity via the microbiota-gut-brain axis in colitis mice. Food Funct 2023; 14:1909-1928. [PMID: 36748225 DOI: 10.1039/d2fo04014j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Large preclinical evidence suggested that colitis was one of the risk factors for depression and probiotics were effective therapeutic agents to prevent the disease. The effect of Lacticaseibacillus rhamnosus Fmb14 on colitis-related depression-like behavior and its possible mechanisms were investigated. One week of DSS exposure led to the following changes in male C57BL/6N mice: a reduction in the movement distance from 2218 to 1299 cm, time in central areas from 23.6 s to 11.5 s, and time in the bright box from 217 s to 103 s, which were restored to 1816 cm, 18.4 s, and 181 s, respectively, with preadministration of Fmb14 for 8 weeks. All improvements provided by Fmb14 indicated a remarkable protective effect on depression-like behavior. Fmb14 first worked to repair intestinal barrier damage and the inflammatory response in the colon through ZO1 and Ocln enhancement and IL-1β, NF-κB and IL-6 reduction, respectively. Second, dysbiosis of the gut microbiota was modulated by Fmb14, including reduction of Akkermansia (18.9% to 5.4%), Mucispirillum (0.6% to 0.1%) and Bifidobacterium (0.32% to 0.03%). Fmb14 supplementation ameliorates the brain inflammatory response via IL-18 and NF-κB reduction and improves the blood-brain barrier via increased levels of ZO1 and Ocln. Moreover, brain activity was facilitated by an increase in BDNF and dopamine and the downregulation of GABA in the Fmb14 group. As a consequence of the modulatory effect on the dysfunction of neurotransmitters and neuroinflammation, Fmb14 prevents neurodegeneration by inhibiting neuronal apoptosis and Nissl edema. In addition, the correlation analysis further demonstrated the preventative effect of Fmb14 on depression-like behavior through the microbiota-gut-brain axis. Together, these findings demonstrated the important role of Fmb14 in biological signal transduction over the microbiota-gut-brain axis to improve mood disorders.
Collapse
Affiliation(s)
- Hongyuan Zhao
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Xiaoyu Chen
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Li Zhang
- Institute of Vegetable, Gansu Academy of Agricultural Sciences, Lanzhou 730070, China
| | - Chao Tang
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Fanqiang Meng
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Libang Zhou
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Ping Zhu
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Zhaoxin Lu
- College of Food Science & Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yingjian Lu
- College of Food Science & Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China.
| |
Collapse
|
41
|
Deng S, Cai K, Pei C, Zhang X, Xiao X, Chen Y, Chen Y, Liang R, Chen Y, Li P, Xie Z, Liao Q. 16S rRNA and Metagenomics Combined with UPLC-Q/TOF-MS Metabolomics Analysis Reveals the Potential Mechanism of Radix Astragali Against Hyperuricemia in Mice. Drug Des Devel Ther 2023; 17:1371-1386. [PMID: 37181826 PMCID: PMC10171225 DOI: 10.2147/dddt.s407983] [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: 02/23/2023] [Accepted: 04/29/2023] [Indexed: 05/16/2023] Open
Abstract
Purpose This study aimed to investigate the underlying treatment mechanism of Radix Astragali (RA) in hyperuricemia from the perspective of microbiota and metabolomics. Methods We used potassium oxyazinate (PO) to induce hyperuricemia mice, and we determined serum alanine aminotransferase/aspartate aminotransferase (ALT/AST), xanthine oxidase (XOD), creatinine (CRE), uric acid (UA), blood urea nitrogen (BUN) levels, liver XOD levels and assessed the kidney tissue histopathology. The therapeutic mechanism of RA in hyperuricemic mice was studied by 16S rRNA, metagenomic sequencing and metabolomics. Results Our research showed that RA has therapeutic effect in hyperuricemia mice, such as slow the weight loss, repair kidney damage, and downregulate serum UA, XOD, CRE, ALT/AST, BUN, and liver XOD levels. RA restored the disturbance structure of the microbiota in hyperuricemia mice by increasing the relative abundances of beneficial bacteria (Lactobacillaceae and Lactobacillus murine) but decreasing the relative abundances of pathogenic bacteria (Prevotellaceae, Rikenellaceae and Bacteroidaceae). Meanwhile, we found that RA directly regulated the metabolic pathway (such as linoleic acid metabolism and glycerophospholipid metabolism) and indirectly regulated bile acid metabolism by mediating microbiota to ameliorate metabolic disorders. Subsequently, there was a robust correlation between specific microbiota, metabolites and the disease index. Conclusion The ability of RA to protect mice against hyperuricemia is strongly linked to the microbiome-metabolite axis, which would provide evidence for RA as a medicine to prevent or treat hyperuricemia.
Collapse
Affiliation(s)
- Song Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Kaiwei Cai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Chaoying Pei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Xingyuan Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Xiaoyi Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Ye Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Ying Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Rongyao Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Yanlong Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Pei Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
- Zhiyong Xie, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, 510006, People’s Republic of China, Tel/Fax +86 075523260207, Email
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
- Correspondence: Qiongfeng Liao, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China, Tel/Fax +86 02039358081, Email
| |
Collapse
|