1
|
Song S, Shi K, Fan M, Wen X, Li J, Guo Y, Lou Y, Chen F, Wang J, Huang L, Wen C, Shao T. Clostridium butyricum and its metabolites regulate macrophage polarization through miR-146a to antagonize gouty arthritis. J Adv Res 2025:S2090-1232(25)00354-6. [PMID: 40398744 DOI: 10.1016/j.jare.2025.05.036] [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: 08/15/2024] [Revised: 03/30/2025] [Accepted: 05/18/2025] [Indexed: 05/23/2025] Open
Abstract
INTRODUCTION Gut microbiota modulation has recently been identified as a prospective avenue for the exploration of novel therapeutic strategies for the management of gout. Nevertheless, the application of a single specific strain or bacterial metabolite for gout intervention has rarely been explored and the underlying regulatory mechanism remains elusive. OBJECTIVES To ascertain the potential role and the molecular mechanism of Clostridium butyricum and butyrate in the management of gouty arthritis. METHODS A Uox-KO mouse model of gouty arthritis was developed and the composition of the gut microbiota was analyzed. C. butyricum and butyrate were supplemented to assess functional recovery and intestinal homeostasis. NanoString analysis identified miRNA variations. GC/MS measured butyric acid levels and qPCR detected the abundance of butyrate-producing enzymes and bacteria. Flow cytometry analyzed macrophage polarization and ELISA measured pro-inflammatory cytokine production. Agomir and antagomir were transfected and dual-luciferase reporter assay was adapted for validation of miRNA target binding. siRNA and rescue experiments were performed to validate the role of SOCS7 in macrophage polarization. In addition, a cohort of patients with gouty arthritis were assembled for the purpose of validating the molecular mechanism. RESULTS The results of our study demonstrated that a reduction of butyrate levels, resulting from a deficiency of butyrate-producing bacteria, leads to aberrant miR-146a expression. This, in turn, induces an imbalance in macrophage polarization and the onset of gouty arthritis. The administration of C. butyricum and butyrate demonstrated considerable anti-inflammatory efficacy by restoring intestinal homeostasis, modulating miR-146a expression, and skewing macrophage polarization. The SOCS7/JAK2-STAT3 signaling pathway was identified as a pivotal mediator in the skewing of macrophage polarization induced by miR-146a. CONCLUSION Our findings enrich the understanding of the regulatory mechanisms underlying macrophage polarization in gouty arthritis and highlight the potential applications of probiotics and their metabolites in clinical gout treatment.
Collapse
Affiliation(s)
- Siyue Song
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Kaiyue Shi
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Moqi Fan
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xianghui Wen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiatao Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yining Guo
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yu Lou
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Fusen Chen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jialu Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lin Huang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Innovative Basic Research in Autoimmune Diseases in Medicine, Hangzhou 310053, China
| | - Chengping Wen
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Innovative Basic Research in Autoimmune Diseases in Medicine, Hangzhou 310053, China.
| | - Tiejuan Shao
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Center for Innovative Basic Research in Autoimmune Diseases in Medicine, Hangzhou 310053, China.
| |
Collapse
|
2
|
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
|
3
|
Huang L, Chen C, Meng J, Yan Q, Luo G, Sha S, Xing Y, Liu C, Xu M, Zhao L, Guo S, Wu X, Chen H, Ma J, You W, Zhang Y, Guo R, Li S, Yao X, Ma W, Kong X, Zhou P, Sun W. Metagenome-Based Characterization of the Gut Virome Signatures in Patients With Gout. J Med Virol 2025; 97:e70336. [PMID: 40207877 DOI: 10.1002/jmv.70336] [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: 12/02/2024] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
The gut microbiome has been implicated in the development of autoimmune diseases, including gout. However, the role of the gut virome in gout pathogenesis remains underexplored. We employed a reference-dependent virome approach to analyze fecal metagenomic data from 102 gout patients (77 in the discovery cohort and 25 in the validation cohort) and 86 healthy controls (HCs) (63 and 23 in each cohort). A subset of gout patients in the discovery cohort provided longitudinal samples at Weeks 2, 4, and 24. Our analysis revealed significant alterations in the gut virome of gout patients, including reduced viral richness and shifts in viral family composition. Notably, Siphoviridae, Myoviridae, and Podoviridae were depleted, while Quimbyviridae, Retroviridae, and Schitoviridae were enriched in gout patients. We identified 359 viral operational taxonomic units (vOTUs) associated with gout. Enriched vOTUs in gout patients predominantly consisted of Fusobacteriaceae, Bacteroidaceae, and Selenomonadaceae phages, while control-enriched vOTUs included Ruminococcaceae, Oscillospiraceae, and Enterobacteriaceae phages. Longitudinal analysis revealed that a substantial proportion of these virome signatures remained stable over 6 months. Functional profiling highlighted the enrichment of viral auxiliary metabolic genes, suggesting potential metabolic interactions between viruses and host bacteria. Notably, gut virome signatures effectively discriminated gout patients from HCs, with high classification performance in the validation cohort. This study provides the first comprehensive characterization of the gut virome in gout, revealing its potential role in disease pathogenesis and highlighting virome-based signatures as promising biomarkers for gout diagnosis and future therapeutic strategies.
Collapse
Affiliation(s)
- Liansha Huang
- Department of Reproductive Health, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Changming Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | | | - Qiulong Yan
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
- Central Hospital of Loudi, Loudi, China
| | - Guangbin Luo
- Centre for Translational Medicine, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Shanshan Sha
- Department of Microbiology, Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yida Xing
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Changyan Liu
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mingxi Xu
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lin Zhao
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shumin Guo
- Department of Reproductive Health, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiaoling Wu
- Department of Reproductive Health, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Huiling Chen
- Department of Reproductive Health, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jie Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei You
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yan Zhang
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ruochun Guo
- The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, China
| | | | - Xueming Yao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wukai Ma
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaodan Kong
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Peng Zhou
- Department of Acupuncture, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Wen Sun
- Centre for Translational Medicine, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Martínez-Nava GA, Altamirano-Molina E, Vázquez-Mellado J, Casimiro-Soriguer CS, Dopazo J, Lozada-Pérez C, Herrera-López B, Martínez-Gómez LE, Martínez-Armenta C, Guido-Gómora DL, Valle-Gutiérrez S, Suarez-Ahedo C, Camacho-Rea MDC, Martínez-García M, Gutiérrez-Esparza G, Amezcua-Guerra LM, Zamudio-Cuevas Y, Martínez-Flores K, Fernández-Torres J, Burguete-García AI, Orbe-Orihuela YC, Lagunas-Martínez A, Méndez-Salazar EO, Francisco-Balderas A, Palacios-González B, Pineda C, López-Reyes A. Metatranscriptomic analysis reveals gut microbiome bacterial genes in pyruvate and amino acid metabolism associated with hyperuricemia and gout in humans. Sci Rep 2025; 15:9981. [PMID: 40121243 PMCID: PMC11929762 DOI: 10.1038/s41598-025-93899-1] [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: 11/07/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Several pathologies with metabolic origin, such as hyperuricemia and gout, have been associated with the gut microbiota taxonomic profile. However, there is no evidence of which bacterial genes are being expressed in the gut microbiome, and of their potential effects on hyperuricemia and gout. We sequenced the RNA of 26 fecal samples from 10 healthy normouricemic controls, 10 with asymptomatic hyperuricemia (AH), and six gout patients. The coding sequences were mapped to KEGG orthologues (KO). We compared the expression levels using generalized linear models and validated the expression of four KO in a larger sample by qRT-PCR. A distinct genetic expression pattern was identified among groups. AH individuals and gout patients showed an over-expression of KOs mainly related to pyruvate metabolism (Log2foldchange > 23, p-adj ≤ 3.56 × 10- 9), the pentose pathway (Log2foldchange > 24, p-adj < 1.10 × 10-12) and purine metabolism (Log2foldchange > 22, p-adj < 1.25 × 10- 7). AH subjects had lower expression of KO related to glycine metabolism (Log2foldchange=-18, p-adj < 1.72 × 10-6) than controls. Gout patients had lower expression (Log2foldchange=-22.42, p-adj < 3.31 × 10- 16) of a KO involved in phenylalanine biosynthesis, in comparison to controls and AH subjects. The over-expression seen for the KO related to pyruvate metabolism and the pentose pathway in gout patients´ microbiome was validated. There is a differential gene expression pattern in the gut microbiome of normouricemic individuals, AH subjects and gout patients. These differences are mainly located in metabolic pathways involved in acetate precursors and bioavailability of amino acids.
Collapse
Affiliation(s)
- Gabriela Angélica Martínez-Nava
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Efren Altamirano-Molina
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Alcaldía Miguel Hidalgo, C.P. 11340, Ciudad de México, Mexico
- Servicio de Reumatología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, C.P. 4389, CDMX, Mexico
| | - Janitzia Vázquez-Mellado
- Servicio de Reumatología, Hospital General de México Eduardo Liceaga, Dr. Balmis 148, Doctores, Cuauhtémoc, C.P. 06720, CDMX, Mexico
| | - Carlos S Casimiro-Soriguer
- Plataforma de Medicina Computacional, Fundación Progreso y Salud (FPS), CDCA, Hospital Vírgen del Rocio, 41013, Sevilla, España
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Sevilla, 41013, Sevilla, España
| | - Joaquín Dopazo
- Plataforma de Medicina Computacional, Fundación Progreso y Salud (FPS), CDCA, Hospital Vírgen del Rocio, 41013, Sevilla, España
- Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Sevilla, 41013, Sevilla, España
| | - Carlos Lozada-Pérez
- Servicio de Reumatología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, C.P. 4389, CDMX, Mexico
| | - Brígida Herrera-López
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Laura Edith Martínez-Gómez
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Carlos Martínez-Armenta
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Dafne Lissete Guido-Gómora
- Servicio de reconstrucción articular de cadera y rodilla, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, CDMX, C.P. 14389, Mexico
| | - Sarahí Valle-Gutiérrez
- Universidad Autónoma Metropolitana Iztapalapa, Av. Ferrocarril San Rafael Atlixco, Núm. 186, Col. Leyes de Reforma 1 A Sección, Alcaldía Iztapalapa, Tlalpan, C.P. 09310, CDMX, Mexico
| | - Carlos Suarez-Ahedo
- Servicio de reconstrucción articular de cadera y rodilla, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, CDMX, C.P. 14389, Mexico
- Departamento de ortopedia, oficina de cirugía, Hospital Médica Sur, Puente de Piedra No. 150, Col. Toriello Guerra, C.P.14050, CDMX, Mexico
| | - María Del Carmen Camacho-Rea
- Departamento de Nutrición Animal, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, Mexico
| | - Mireya Martínez-García
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Belisario Domínguez Secc 16, Tlalpan, 14080, CDMX, Mexico
| | - Guadalupe Gutiérrez-Esparza
- Programa Investigador para México de la SECIHTI, Secretaría de Ciencias, Humanidades, Tecnología e Innovación, Ciudad de México, Avenida Insurgentes Sur 1582, Crédito Constructor, CDMX, Mexico
- Servicios de Diagnóstico y Tratamiento, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Belisario Domínguez Secc 16, Tlalpan, 14080, CDMX, Mexico
| | - Luis M Amezcua-Guerra
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Belisario Domínguez Secc 16, Tlalpan, 14080, CDMX, Mexico
| | - Yessica Zamudio-Cuevas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Karina Martínez-Flores
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Javier Fernández-Torres
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Ana I Burguete-García
- GID Microbiota y Epidemiologia Genética, Instituto Nacional de Salud Pública, Universidad No. 655 Colonia Santa María Ahuacatitlán, Cuernavaca, Morelos, Mexico
| | - Yaneth Citlalli Orbe-Orihuela
- GID Microbiota y Epidemiologia Genética, Instituto Nacional de Salud Pública, Universidad No. 655 Colonia Santa María Ahuacatitlán, Cuernavaca, Morelos, Mexico
| | - Alfredo Lagunas-Martínez
- GID Microbiota y Epidemiologia Genética, Instituto Nacional de Salud Pública, Universidad No. 655 Colonia Santa María Ahuacatitlán, Cuernavaca, Morelos, Mexico
| | | | - Adriana Francisco-Balderas
- Hospital General de Zona No. 71 "Benito Coquet Lagunes", Instituto Mexicano de la Seguridad Social, Av. Salvador Díaz Mirón s/n, Pastora, Floresta, C.P. 91930, Veracruz, Mexico
| | - Berenice Palacios-González
- Laboratorio de Envejecimiento Saludable del INMEGEN en el Centro de Investigación sobre el Envejecimiento, Calz. de los Tenorios 235. Col. Rinconada de las Hadas, Tlalpan, 14330, CDMX, Mexico
- Dirección de Investigación, Instituto Nacional de Medicina Genómica (INMEGEN), Periférico Sur 4809, Tlalpan, México, 14610, Mexico, Mexico
| | - Carlos Pineda
- Dirección General, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, Mexico.
- Laboratorio de Gerociencias, Luis Guillermo Ibarra Ibarra National Rehabilitation Institute, Calz México- Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, Ciudad de México, CDMX, Mexico.
| |
Collapse
|
5
|
Ahn EY, So MW. The pathogenesis of gout. JOURNAL OF RHEUMATIC DISEASES 2025; 32:8-16. [PMID: 39712248 PMCID: PMC11659655 DOI: 10.4078/jrd.2024.0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/19/2024] [Accepted: 10/24/2024] [Indexed: 12/24/2024]
Abstract
Gout is the most common inflammatory arthritis in adults, associated with hyperuricemia and the chronic deposition of monosodium urate (MSU) crystals. Hyperuricemia results from increased production of uric acid and decreased excretion by the kidneys and intestines. Urate excretion is regulated by a group of urate transporters, and decreased renal or intestinal excretion is the primary mechanism of hyperuricemia in most people. Genetic variability in these urate transporters is strongly related to variances in serum urate levels. Not all individuals with hyperuricemia show deposition of MSU crystals or develop gout. The initiation of the inflammatory response to MSU crystals is mainly mediated by the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing protein 3 (NLRP3) inflammasome. The activated NLRP3 inflammasome complex cleaves pro-interleukin-1β (IL-1β) into its active form, IL-1β, which is a key mediator of the inflammatory response in gout. IL-1β leads to the upregulation of cytokines and chemokines, resulting in the recruitment of neutrophils and other immune cells. Neutrophils recruited to the site of inflammation also play a role in resolving inflammation. Aggregated neutrophil extracellular traps (NETs) trap and degrade cytokines and chemokines through NET-bound proteases, promoting the resolution of inflammation. Advanced gout is characterized by tophi, chronic inflammatory responses, and structural joint damage. Tophi are chronic foreign body granuloma-like structures containing collections of MSU crystals encased by inflammatory cells and connective tissue. Tophi are closely related to chronic inflammation and structural damage.
Collapse
Affiliation(s)
- Eun Young Ahn
- Division of Rheumatology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Min Wook So
- Division of Rheumatology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
6
|
La Monica MB, Raub B, Hartshorn S, Gustat AL, Grdic J, Kirby TO, Townsend JR, Sandrock J, Ziegenfuss TN. The effects of AG1® supplementation on the gut microbiome of healthy adults: a randomized, double-blind, placebo-controlled clinical trial. J Int Soc Sports Nutr 2024; 21:2409682. [PMID: 39352252 PMCID: PMC11445888 DOI: 10.1080/15502783.2024.2409682] [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: 02/16/2024] [Accepted: 09/21/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND This study aimed to examine the effect of a commercially available multi-ingredient powder (AG1Ⓡ) on the gut microbiome and assess the impact of AG1Ⓡ on GI tolerability and other clinical safety markers in healthy men and women. METHODS Using a double-blind, randomized, two-arm, placebo-controlled, parallel design, we examined a 4-week daily supplementation regimen of AG1Ⓡ vs. placebo (PL). Fifteen men and 15 women provided stool samples for microbiome analysis, questionnaires for digestive quality of life (DQLQ), and completed visual analog scales (VAS) and Bristol stool charts to assess stool consistency and bowel frequency before and after the 4-week intervention. Participant's blood work (CBC, CMP, and lipid panel) was also assessed before and after the 4-week intervention. Alpha diversity was determined by Shannon and Chao1 index scores and evaluated by a two-way ANOVA, beta diversity in taxonomic abundances and functional pathways was visualized using partial least squares-discriminant analyses and statistically evaluated by PERMANOVA. To identify key biomarkers, specific feature differences in taxonomic relative abundance and normalized functional pathway counts were analyzed by linear discriminant analysis (LDA) effect size (LEfSe). Questionnaires, clinical safety markers, and hemodynamics were evaluated by mixed factorial ANOVAs with repeated measures. This study was registered on clinicaltrials.gov (NCT06181214). RESULTS AG1Ⓡ supplementation enriched two probiotic taxa (Lactobacillus acidophilus and Bifidobacterium bifidum) that likely stem from the probiotics species that exist in the product, as well as L. lactis CH_LC01 and Acetatifactor sp900066565 ASM1486575v1 while reducing Clostridium sp000435835. Regarding community function, AG1Ⓡ showed an enrichment of two functional pathways while diminishing none. Alternatively, the PL enriched six, but diminished five functional pathways. Neither treatment negatively impacted the digestive quality of life via DQLQ, bowel frequency via VAS, or stool consistency via VAS and Bristol. However, there may have been a greater improvement in the DQLQ score (+62.5%, p = 0.058, d = 0.73) after four weeks of AG1Ⓡ supplementation compared to a reduction (-50%) in PL. Furthermore, AG1Ⓡ did not significantly alter clinical safety markers following supplementation providing evidence for its safety profile. CONCLUSIONS AG1Ⓡ can be consumed safely by healthy adults over four weeks with a potential beneficial impact in their digestive symptom quality of life.
Collapse
Affiliation(s)
| | - Betsy Raub
- The Center for Applied Health Sciences, Canfield, OH, USA
| | | | | | - Jodi Grdic
- The Center for Applied Health Sciences, Canfield, OH, USA
| | - Trevor O. Kirby
- AG1, Research, Nutrition, and Innovation, Carson City, NV, USA
| | - Jeremy R. Townsend
- AG1, Research, Nutrition, and Innovation, Carson City, NV, USA
- Concordia University Chicago, Health & Human Performance, River Forest, IL, USA
| | - Jen Sandrock
- The Center for Applied Health Sciences, Canfield, OH, USA
| | | |
Collapse
|
7
|
Zhou L, Zhang Y, Wu S, Kuang Y, Jiang P, Zhu X, Yin K. Type III Secretion System in Intestinal Pathogens and Metabolic Diseases. J Diabetes Res 2024; 2024:4864639. [PMID: 39544522 PMCID: PMC11561183 DOI: 10.1155/2024/4864639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Modern lifestyle changes, especially the consumption of a diet high in salt, sugar, and fat, have contributed to the increasing incidence and prevalence of chronic metabolic diseases such as diabetes, obesity, and gout. Changing lifestyles continuously shape the gut microbiota which is closely related to the occurrence and development of metabolic diseases due to its specificity of composition and structural diversity. A large number of pathogenic bacteria such as Yersinia, Salmonella, Shigella, and pathogenic E. coli in the gut utilize the type III secretion system (T3SS) to help them resist host defenses and cause disease. Although the T3SS is critical for the virulence of many important human pathogens, its relationship with metabolic diseases remains unknown. This article reviews the structure and function of the T3SS, the disruption of intestinal barrier integrity by the T3SS, the changes in intestinal flora containing the T3SS in metabolic diseases, the possible mechanisms of the T3SS affecting metabolic diseases, and the application of the T3SS in the treatment of metabolic diseases. The aim is to provide insights into metabolic diseases targeting the T3SS, thereby serving as a valuable reference for future research on disease diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Yaoyuan Zhang
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510900, China
| | - Shiqi Wu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Yiyu Kuang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Pengfei Jiang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou 510900, China
| |
Collapse
|
8
|
Singh AK, Durairajan SSK, Iyaswamy A, Williams LL. Elucidating the role of gut microbiota dysbiosis in hyperuricemia and gout: Insights and therapeutic strategies. World J Gastroenterol 2024; 30:4404-4410. [PMID: 39494101 PMCID: PMC11525862 DOI: 10.3748/wjg.v30.i40.4404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/16/2024] Open
Abstract
Hyperuricemia (HUA) is a condition associated with a high concentration of uric acid (UA) in the bloodstream and can cause gout and chronic kidney disease. The gut microbiota of patients with gout and HUA is significantly altered compared to that of healthy people. This article focused on the complex interconnection between alterations in the gut microbiota and the development of this disorder. Some studies have suggested that changes in the composition, diversity, and activity of microbes play a key role in establishing and progressing HUA and gout pathogenesis. Therefore, we discussed how the gut microbiota contributes to HUA through purine metabolism, UA excretion, and intestinal inflammatory responses. We examined specific changes in the composition of the gut microbiota associated with gout and HUA, highlighting key bacterial taxa and the metabolic pathways involved. Additionally, we discussed the effect of conventional gout treatments on the gut microbiota composition, along with emerging therapeutic approaches that target the gut microbiome, such as the use of probiotics and prebiotics. We also provided insights into a study regarding the gut microbiota as a possible novel therapeutic intervention for gout treatment and dysbiosis-related diagnosis.
Collapse
Affiliation(s)
- Abhay Kumar Singh
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, TN 610005, India
| | - Siva Sundara Kumar Durairajan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, TN 610005, India
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ashok Iyaswamy
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Leonard L Williams
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| |
Collapse
|
9
|
Wang Z, Xu H, Song X, Chen Z, Wang G, Yang Y, Zhu B, Ai L, Liu C, Zhang Y, Yang Y, Wang C, Xia Y. Revealing Interactions of Gut Microbiota and Metabolite in Confined Environments Using High-Throughput Sequencing and Metabolomic Analysis. Nutrients 2024; 16:2998. [PMID: 39275313 PMCID: PMC11397237 DOI: 10.3390/nu16172998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
A confined environment is a special kind of extreme working environment, and prolonged exposure to it tends to increase psychological stress and trigger rhythmic disorders, emotional abnormalities and other phenomena, thus seriously affecting work efficiency. However, the mechanisms through which confined environments affect human health remain unclear. Therefore, this study simulates a strictly controlled confined environment and employs integrative multi-omics techniques to analyze the alterations in gut microbiota and metabolites of workers under such conditions. The aim is to identify metabolic biomarkers and elucidate the relationship between gut microbiota and metabolites. High-throughput sequencing results showed that a confined environment significantly affects gut microbial composition and clusters subjects' gut microbiota into two enterotypes (Bla and Bi). Differences in abundance of genera Bifidobacterium, Collinsella, Ruminococcus_gnavus_group, Faecalibacterium, Bacteroides, Prevotella and Succinivibronaceae UCG-002 were significant. Untarget metabolomics analyses showed that the confined environment resulted in significant alterations in intestinal metabolites and increased the activity of the body's amino acid metabolism and bile acid metabolism pathways. Among the metabolites that differed after confined environment living, four metabolites such as uric acid and beta-PHENYL-gamma-aminobutyric acid may be potential biomarkers. Further correlation analysis demonstrated a strong association between the composition of the subjects' gut microbiota and these four biomarkers. This study provides valuable reference data for improving the health status of workers in confined environments and facilitates the subsequent proposal of targeted prevention and treatment strategies.
Collapse
Affiliation(s)
- Ziying Wang
- Naval Medical Center, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai 200433, China
| | - Haodan Xu
- Naval Medical Center, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai 200433, China
| | - Xin Song
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zheng Chen
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Guangqiang Wang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yijin Yang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Beiwei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Lianzhong Ai
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Chenxi Liu
- Naval Medical Center, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai 200433, China
| | - Yaxuan Zhang
- Naval Medical Center, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai 200433, China
| | - Yong Yang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Chuan Wang
- Naval Medical Center, Naval Medical University, Shanghai 200433, China
- Key Laboratory of Molecular Neurobiology of Ministry of Education, Shanghai 200433, China
| | - Yongjun Xia
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China
| |
Collapse
|
10
|
Wang H, Zheng Y, Yang M, Wang L, Xu Y, You S, Mao N, Fan J, Ren S. Gut microecology: effective targets for natural products to modulate uric acid metabolism. Front Pharmacol 2024; 15:1446776. [PMID: 39263572 PMCID: PMC11387183 DOI: 10.3389/fphar.2024.1446776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Gut microecology,the complex community consisting of microorganisms and their microenvironments in the gastrointestinal tract, plays a vital role in maintaining overall health and regulating various physiological and pathological processes. Recent studies have highlighted the significant impact of gut microecology on the regulation of uric acid metabolism. Natural products, including monomers, extracts, and traditional Chinese medicine formulations derived from natural sources such as plants, animals, and microorganisms, have also been investigated for their potential role in modulating uric acid metabolism. According to research, The stability of gut microecology is a crucial link for natural products to maintain healthy uric acid metabolism and reduce hyperuricemia-related diseases. Herein, we review the recent advanced evidence revealing the bidirectional regulation between gut microecology and uric acid metabolism. And separately summarize the key evidence of natural extracts and herbal formulations in regulating both aspects. In addition,we elucidated the important mechanisms of natural products in regulating uric acid metabolism and secondary diseases through gut microecology, especially by modulating the composition of gut microbiota, gut mucosal barrier, inflammatory response, purine catalyzation, and associated transporters. This review may offer a novel insight into uric acid and its associated disorders management and highlight a perspective for exploring its potential therapeutic drugs from natural products.
Collapse
Affiliation(s)
- Hui Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yixuan Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengfan Yang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yao Xu
- Chengdu Medical College, Chengdu, China
| | - Siqi You
- Chengdu Medical College, Chengdu, China
| | - Nan Mao
- Chengdu Medical College, Chengdu, China
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Junming Fan
- Chengdu Medical College, Chengdu, China
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Sichong Ren
- Chengdu Medical College, Chengdu, China
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- TCM Preventative Treatment Research Center of Chengdu Medical College, Chengdu, China
| |
Collapse
|
11
|
Li H, Su Q, Fu D, Huang H, Lu Z, Huang C, Chen Y, Tan M, Huang J, Kang Z, Wei Q, Guo X. Alteration of gut microbiome in goslings infected with goose astrovirus. Poult Sci 2024; 103:103869. [PMID: 38909510 PMCID: PMC11253677 DOI: 10.1016/j.psj.2024.103869] [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: 02/08/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 06/25/2024] Open
Abstract
Goose astrovirus (GoAstV) is an emerging avian pathogen that induces gout in goslings with a mortality of up to 50%. Organ damage caused by GoAstV infection was considered the cause of gout, but it is still unclear whether other factors are involved. Human and murine studies have linked the gut microbiome-derived urate and gout, thus we hypothesized that gut microbiome may also play an important role in gout induced by GoAstV infection. This study tested the pathogenicity of our isolated GoAstV genotype 2 strain on goslings, while the appearance of clinical signs, histopathological changes, viral distribution and the blood level of cytokines were monitored for 18 d postinfection (dpi). The dynamics in the gut microbiome were profiled by 16S sequencing and then correlated with GoAstV infection. Results showed that this study successfully developed an experimental infection model for studying the pathogenicity of the GoAstV infection which induces typical symptoms of gout. GoAstV infection significantly altered the gut microbiome of goslings with the enrichment of potential proinflammatory bacteria and depletion of beneficial bacteria that can produce short-chain fatty acids. More importantly, the microbial pathway involved in urate production was significantly increased in goslings infected with GoAstV, suggesting that gut microbiome-derived urate may also contribute to the gout symptoms. Overall, this study demonstrated the role of gut microbiome in the pathogenesis of GoAstV infection, highlighting the potential of gut microbiome-based therapeutics against gout symptoms.
Collapse
Affiliation(s)
- Haiqin Li
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi 330200, China
| | - Qi Su
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Duanfeng Fu
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Haoyu Huang
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Zhihua Lu
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Cheng Huang
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yunfeng Chen
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Meifang Tan
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi 330200, China
| | - Jiangnan Huang
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi 330200, China
| | - Zhaofeng Kang
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi 330200, China
| | - Qipeng Wei
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang, Jiangxi 330200, China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China.
| |
Collapse
|
12
|
Nkeck JR, Tchuisseu-Kwangoua AL, Pelda A, Tamko WC, Hamadjoda S, Essama DB, Fojo B, Niasse M, Diallo S, Ngandeu-Singwé M. Current Approaches to Prevent or Reverse Microbiome Dysbiosis in Chronic Inflammatory Rheumatic Diseases. Mediterr J Rheumatol 2024; 35:220-233. [PMID: 39211023 PMCID: PMC11350408 DOI: 10.31138/mjr.240224.cap] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 09/04/2024] Open
Abstract
Advances in knowledge of the microbiome and its relationship with the immune system have led to a better understanding of the pathogenesis of chronic inflammatory rheumatic diseases (CIRD). Indeed, the microbiome dysbiosis now occupies a particular place with implications for the determinism and clinical expression of CIRD, as well as the therapeutic response of affected patients. Several approaches exist to limit the impact of the microbiome during CIRD. This review aimed to present current strategies to prevent or reverse microbiome dysbiosis based on existing knowledge, in order to provide practical information to healthcare professionals treating patients suffering from CIRD.
Collapse
Affiliation(s)
- Jan René Nkeck
- Yaoundé Rheumatology Research Team, Yaoundé, Cameroon
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Ange Larissa Tchuisseu-Kwangoua
- Yaoundé Rheumatology Research Team, Yaoundé, Cameroon
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Adeline Pelda
- Yaoundé Rheumatology Research Team, Yaoundé, Cameroon
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- Rheumatology Unit, Yaoundé Central Hospital, Yaoundé, Cameroon
| | - Wilson Chia Tamko
- Yaoundé Rheumatology Research Team, Yaoundé, Cameroon
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- Rheumatology Unit, Yaoundé Central Hospital, Yaoundé, Cameroon
| | - Saquinatou Hamadjoda
- Yaoundé Rheumatology Research Team, Yaoundé, Cameroon
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- Rheumatology Unit, Yaoundé Central Hospital, Yaoundé, Cameroon
| | - Doris Bibi Essama
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Baudelaire Fojo
- Yaoundé Rheumatology Research Team, Yaoundé, Cameroon
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- Rheumatology Unit, Yaoundé Central Hospital, Yaoundé, Cameroon
| | - Moustapha Niasse
- Department of Rheumatology, Dantec Teaching Hospital, Cheikh Anta Diop University, Dakar, Senegal
| | - Saïdou Diallo
- Department of Rheumatology, Dantec Teaching Hospital, Cheikh Anta Diop University, Dakar, Senegal
| | - Madeleine Ngandeu-Singwé
- Yaoundé Rheumatology Research Team, Yaoundé, Cameroon
- Department of Internal Medicine and Specialties, Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
- Rheumatology Unit, Yaoundé Central Hospital, Yaoundé, Cameroon
| |
Collapse
|
13
|
Zeng J, Li Y, Zou Y, Yang Y, Yang T, Zhou Y. Intestinal toxicity alleviation and efficacy potentiation through therapeutic administration of Lactobacillus paracasei GY-1 in the treatment of gout flares with colchicine. Food Funct 2024; 15:1671-1688. [PMID: 38251779 DOI: 10.1039/d3fo04858f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Gout flares have emerged as a significant public health concern. Colchicine (COL) is a first-line and standard drug for treating gout flares. However, its clinical use is limited due to various adverse effects. Besides, COL fails to adequately meet the needs of patients, particularly young patients. In this study, we investigate the therapeutic administration of Lactobacillus paracasei GY-1 (GY-1) to overcome the limitations of COL. Our results demonstrate that GY-1 attenuates COL toxicity in terms of body weight loss, decreased feed intake, mortality, reduced locomotor activity, colon shortening, increased oxidative stress, histological damage, and impaired gut permeability. Meanwhile, we demonstrate that GY-1 enhances the therapeutic effect for gout flares when combined with COL, as evidenced by the reduction in paw swelling, decreased levels of proinflammatory cytokines including IL-1β and TNF-α, and an increase in the anti-inflammatory cytokine IL-10. Additionally, the absolute quantification of the gut microbiota shows that GY-1 restores the gut microbiota imbalance caused by COL. Furthermore, GY-1 reduces the abundance of 4 Alistipes species and 6 Porphyromonadaceae species, which may be responsible for toxicity alleviation. At the same time, GY-1 increases the abundance of Bacteroides sartorii and Enterococcus sp., which may contribute to its therapeutic efficacy. This study demonstrates the feasibility of developing probiotic-based adjuvant therapy or bacteriotherapy for treating gout flares. To our knowledge, GY-1 is the first probiotic that could be used as an alternative synergetic agent with COL for the therapeutic treatment of gout flares.
Collapse
Affiliation(s)
- Jiaqi Zeng
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Yan Li
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Yizhi Zou
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Ying Yang
- Department of Public Health, School of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi 545005, China
| | - Tingting Yang
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Yizhuang Zhou
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, Guilin, Guangxi 541199, China.
| |
Collapse
|
14
|
Han J, Li M, Li X, Liu C, Li XL, Wang K, Qiao R, Yang F, Han X, Li XJ. Effects of microbes in pig farms on occupational exposed persons and the environment. AMB Express 2023; 13:136. [PMID: 38032532 PMCID: PMC10689614 DOI: 10.1186/s13568-023-01631-x] [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: 06/09/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
In terms of pig farming, pig gut microbes have a significant effect on farmers and the farm environment. However, it is still unclear which microbial composition is more likely to contribute to this effect. This study collected a total of 136 samples, including pigs' faeces samples, farmers' faeces samples, samples from individuals who had no contact with any type of farm animal (referred to as 'non-exposed' persons), and environmental dust samples (collected from inside and outside pig houses and the farm) from two pig farms, pig farm A and pig farm B. Whereafter, 16S rRNA sequencing and taxonomic composition analysis were performed. According to the study, compared to non-exposed persons, pig farmers had a significantly higher abundance of 7 genera. In addition, the farmers were grouped according to the duration of their occupational exposure, and it was shown that 4 genera, including Turicibacter, Terrisporobacter, and Clostridium_sensu_stricto_1, exhibited a rise in more frequent contact with pigs. As compared to outside the pig house, the environmental dust has a greater concentration of the 3 bacteria mentioned before. Therefore, these 3 microbes can be considered as co-occurring microbes that may exist both in humans and the environment. Also, the 3 co-occurring microbes are involved in the fermentation and production of short-chain fatty acids and their effectiveness decreased as distance from the farm increased. This study shows that the 3 microbes where pig farmers co-occur with the environment come from pig farms, which provides fresh ideas for preventing the spread of microbial aerosols in pig farms and reducing pollution.
Collapse
Affiliation(s)
- Jinyi Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Mengyu Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xin Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Chuang Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xiu-Ling Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Kejun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Ruimin Qiao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Feng Yang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xuelei Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
| | - Xin-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
- Sanya Institute, Hainan Academy of Agricultural Science, Sanya, China.
| |
Collapse
|
15
|
Chen C, Zhang Y, Yao X, Yan Q, Li S, Zhong Q, Liu Z, Tang F, Liu C, Li H, Zhu D, Lan W, Ling Y, Lu D, Xu H, Ning Q, Wang Y, Jiang Z, Zhang Q, Gu G, Sun L, Wang N, Wang G, Zhang A, Ullah H, Sun W, Ma W. Characterizations of the multi-kingdom gut microbiota in Chinese patients with gouty arthritis. BMC Microbiol 2023; 23:363. [PMID: 38001408 PMCID: PMC10668524 DOI: 10.1186/s12866-023-03097-0] [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: 06/15/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
OBJECTIVE The gut microbial composition has been linked to metabolic and autoimmune diseases, including arthritis. However, there is a dearth of knowledge on the gut bacteriome, mycobiome, and virome in patients with gouty arthritis (GA). METHODS We conducted a comprehensive analysis of the multi-kingdom gut microbiome of 26 GA patients and 28 healthy controls, using whole-metagenome shotgun sequencing of their stool samples. RESULTS Profound alterations were observed in the gut bacteriome, mycobiome, and virome of GA patients. We identified 1,117 differentially abundant bacterial species, 23 fungal species, and 4,115 viral operational taxonomic units (vOTUs). GA-enriched bacteria included Escherichia coli_D GENOME144544, Bifidobacterium infantis GENOME095938, Blautia_A wexlerae GENOME096067, and Klebsiella pneumoniae GENOME147598, while control-enriched bacteria comprised Faecalibacterium prausnitzii_G GENOME147678, Agathobacter rectalis GENOME143712, and Bacteroides_A plebeius_A GENOME239725. GA-enriched fungi included opportunistic pathogens like Cryptococcus neoformans GCA_011057565, Candida parapsilosis GCA_000182765, and Malassezia spp., while control-enriched fungi featured several Hortaea werneckii subclades and Aspergillus fumigatus GCA_000002655. GA-enriched vOTUs mainly attributed to Siphoviridae, Myoviridae, Podoviridae, and Microviridae, whereas control-enriched vOTUs spanned 13 families, including Siphoviridae, Myoviridae, Podoviridae, Quimbyviridae, Phycodnaviridae, and crAss-like. A co-abundance network revealed intricate interactions among these multi-kingdom signatures, signifying their collective influence on the disease. Furthermore, these microbial signatures demonstrated the potential to effectively discriminate between patients and controls, highlighting their diagnostic utility. CONCLUSIONS This study yields crucial insights into the characteristics of the GA microbiota that may inform future mechanistic and therapeutic investigations.
Collapse
Affiliation(s)
- Changming Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, China
| | - Xueming Yao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiulong Yan
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | | | - Qin Zhong
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhengqi Liu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Fang Tang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Can Liu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hufan Li
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dan Zhu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Weiya Lan
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yi Ling
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Daomin Lu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hui Xu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiaoyi Ning
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ying Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zong Jiang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiongyu Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Guangzhao Gu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Liping Sun
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Nan Wang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Guangyang Wang
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | | | - Hayan Ullah
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Wen Sun
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China.
| | - Wukai Ma
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.
| |
Collapse
|
16
|
Yang L, Liu X, Yan S, Xiong S, Bai X, Yan Y. Highly expressed long non-coding RNA SNHG14 activated MSU-induced inflammatory response in acute gout arthritis through targeting miR-223-3p. Int J Rheum Dis 2023; 26:2233-2239. [PMID: 37715329 DOI: 10.1111/1756-185x.14919] [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: 05/31/2022] [Revised: 08/22/2023] [Accepted: 09/03/2023] [Indexed: 09/17/2023]
Abstract
AIM According to reports, long non-coding RNAs (lncRNAs) are involved in the regulation of many inflammatory diseases. Here, our main purpose was to ascertain the expression data of lncRNA SNHG14 in acute gouty arthritis (AGA) and to explore its possible mechanism in the regulation of AGA. METHOD Reverse transcription quantitative polymerase chain reaction technology was supplied to detect the lncRNA SNHG14 expression. A receiver operating characteristics curve was drawn to estimate the accuracy of lncRNA SNHG14 in AGA diagnosis. An in vitro AGA cell model was constructed by inducing THP-1 cells with monosodium urate (MSU). The concentrations of inflammatory factors such as interleukin-1β, interleukin-6, and tumor necrosis factor-α were measured by enzyme-linked immunosorbent assay. The luciferase reporter gene was used to verify the relationship between lncRNA SNHG14 and miR-223-3p. RESULTS In clinical analysis, the levels of serum lncRNA SNHG14 in AGA patients were significantly higher than those in the control group. Abnormally elevated lncRNA SNHG14 has high sensitivity and specificity for AGA diagnosis. In in vitro cell experiments, silencing lncRNA SNHG14 inhibited the inflammatory response of THP-1 cells stimulated by MSU, and the luciferase reporter gene proved that lncRNA SNHG14 could bind to miR-223-3p. In addition, the level of miR-223-3p declined in AGA patients and the AGA cell model. Overexpression of miR-223-3p is helpful to alleviate an MSU-induced inflammatory response. CONCLUSION In the AGA cell model, lncRNA SNHG14, as an miR-223-3p sponge, induces a cellular inflammatory response by controlling the level of miR-223-3p, so aggravating the disease progress of AGA.
Collapse
Affiliation(s)
- Lu Yang
- Clinical Research Center, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Xiaochuan Liu
- Clinical Research Center, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Shuyi Yan
- Clinical Research Center, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Shili Xiong
- Clinical Research Center, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Xiaosong Bai
- Department of Clinical Laboratory, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Ying Yan
- Shanghai Baoshan Center for Disease Control and Prevention, Shanghai, China
| |
Collapse
|
17
|
Terkeltaub R. Emerging Urate-Lowering Drugs and Pharmacologic Treatment Strategies for Gout: A Narrative Review. Drugs 2023; 83:1501-1521. [PMID: 37819612 DOI: 10.1007/s40265-023-01944-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/13/2023]
Abstract
Hyperuricemia with consequent monosodium urate crystal deposition leads to gout, characterized by painful, incapacitating inflammatory arthritis flares that are also associated with increased cardiovascular event and related mortality risk. This narrative review focuses on emerging pharmacologic urate-lowering treatment (ULT) and management strategies in gout. Undertreated, gout can progress to palpable tophi and joint damage. In oral ULT clinical trials, target serum urate of < 6.0 mg/dL can be achieved in ~ 80-90% of subjects, with flare burden reduction by 1-2 years. However, real-world ULT results are far less successful, due to both singular patient nonadherence and prescriber undertreatment, particularly in primary care, where most patients are managed. Multiple dose titrations commonly needed to optimize first-line allopurinol ULT monotherapy, and substantial potential toxicities and other limitations of approved, marketed oral monotherapy ULT drugs, promote hyperuricemia undertreatment. Common gout comorbidities with associated increased mortality (e.g., moderate-severe chronic kidney disease [CKD], type 2 diabetes, hypertension, atherosclerosis, heart failure) heighten ULT treatment complexity and emphasize unmet needs for better and more rapid clinically significant outcomes, including attenuated gout flare burden. The gout drug armamentarium will be expanded by integrating sodium-glucose cotransporter-2 (SGLT2) inhibitors with uricosuric and anti-inflammatory properties as well as clinically indicated antidiabetic, nephroprotective, and/or cardioprotective effects. The broad ULT developmental pipeline is loaded with multiple uricosurics that selectively target uric acid transporter 1 (URAT1). Evolving ULT approaches include administering selected gut anaerobic purine degrading bacteria (PDB), modulating intestinal urate transport, and employing liver-targeted xanthine oxidoreductase mRNA knockdown. Last, emerging measures to decrease the immunogenicity of systemically administered recombinant uricases should simplify treatment regimens and further improve outcomes in managing the most severe gout phenotypes.
Collapse
Affiliation(s)
- Robert Terkeltaub
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California, 9500 Gilman Drive, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
18
|
Duan Z, Fu J, Zhang F, Cai Y, Wu G, Ma W, Zhou H, He Y. The association between BMI and serum uric acid is partially mediated by gut microbiota. Microbiol Spectr 2023; 11:e0114023. [PMID: 37747198 PMCID: PMC10581133 DOI: 10.1128/spectrum.01140-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/20/2023] [Indexed: 09/26/2023] Open
Abstract
Obesity is a risk factor for the development of hyperuricemia, both of which were related to gut microbiota. However, whether alterations in the gut microbiota lie in the pathways mediating obesity's effects on hyperuricemia is less clear. Body mass index (BMI) and serum uric acid (SUA) were separately important indicators of obesity and hyperuricemia. Our study aims to investigate whether BMI-related gut microbiota characteristics would mediate the association between BMI and SUA levels. A total of 6,280 participants from Guangdong Gut Microbiome Project were included in this study. Stool samples were collected for 16S rRNA gene sequencing. The results revealed that BMI was significantly and positively associated with SUA. Meanwhile, BMI was significantly associated with the abundance of 102 gut microbial genera, 16 of which were also significantly associated with SUA. The mediation analysis revealed that the association between BMI and SUA was partially mediated by the abundance of Proteobacteria (proportion mediated: 0.94%, P < 0.05). At the genus level, 25 bacterial genera, including Ralstonia, Oscillospira, Faecalibacterium, etc., could also partially mediate the association of BMI with SUA (the highest proportion is mediated by Ralstonia, proportion mediated: 2.76%, P < 0.05). This study provided evidence for the associations among BMI, gut microbiota, and SUA, and the mediation analysis suggested that the association of BMI with SUA was partially mediated by the gut microbiota. IMPORTANCE Using 16S rRNA sequencing analysis, local interpretable machine learning technique analysis and mediation analysis were used to explore the association between BMI with SUA, and the mediating effects of gut microbial dysbiosis in the association were investigated.
Collapse
Affiliation(s)
- Zhuo Duan
- Department of Laboratory Medicine, Microbiome Medicine Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingxiang Fu
- Department of Laboratory Medicine, Microbiome Medicine Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Zhang
- Department of Laboratory Medicine, Microbiome Medicine Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yijia Cai
- Department of Laboratory Medicine, Microbiome Medicine Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangyan Wu
- Department of Laboratory Medicine, Microbiome Medicine Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjun Ma
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Centre for Disease Control and Prevention, Guangzhou, Guangdong, China
| | - Hongwei Zhou
- Department of Laboratory Medicine, Microbiome Medicine Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan He
- Department of Laboratory Medicine, Microbiome Medicine Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Saini N, Aamir M, Singh VK, Deepak B, Mona S. Unveiling the microbial diversity and functional dynamics of Shiv Kund, Sohna hot spring, India through a shotgun metagenomics approach. Arch Microbiol 2023; 205:323. [PMID: 37651004 DOI: 10.1007/s00203-023-03664-z] [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: 05/03/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
In this research, we examined the microbial diversity in Sohna hot spring, Haryana, India using shotgun metagenome sequencing based on the Illumina Hiseq 4000 sequencing technology. The raw sequence data from metagenomic paired-end libraries were analysed for taxonomic classification, diversity, and functional annotation using MG-RAST online server. The results showed the presence of total of 57 phyla, 931 genera, and 2068 species, predominantly occupied by Moraxellaceae (Gammaproteobacteria). However, at the species level, we reported the presence of some representative pathogenic taxa, such as Acinetobacter baumannii and Moraxella osloensis. The functional annotation predicted at various levels based on SEED-based subsystem, KEGG ortholog identity (KO), Cluster of Orthologous Groups (COGs) database identified the predominance of genes associated with primary and secondary metabolism along with a crucial role in environmental and genetic signals, cellular communication, and cell signalling. Comparative Genome Analysis (CGA) using The Pathosystem Resource Integration Centre (PATRIC) tool based on genome annotation and assembly of the metagenomic libraries for representative taxon Acinetobacter baumannii (NCBI tax id:470) characterized the reads with a unique genome identifier of 470.20380 (A. baumannii DDLJ4) which is evolutionary closer to A. baumannii ATCC 470.17978 400667.7. In addition, the CARD database results about the presence of potential AMR pathotypes and the prevalence of adeABC, adeIJK, abeM gene-specific clusters that function as multidrug efflux pumps. Overall, the results provided a comprehensive insight into virulence and anti-microbial resistance mechanism and could be useful for developing potential drug targets against the possible AMR pathotypes.
Collapse
Affiliation(s)
- Neha Saini
- Department of Environmental Science and Engineering, Guru Jambheshwar University of Science and Technology, Hisar, India
| | - Mohd Aamir
- Division of Plant Pathology, ICAR-Indian Council of Agricultural Research, Pusa Campus, New Delhi, India
| | - Vinay Kumar Singh
- Centre for Bioinformatics, School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Bansal Deepak
- Department of Environmental Science and Engineering, Guru Jambheshwar University of Science and Technology, Hisar, India
| | - Sharma Mona
- Department of Environmental Studies, School of Interdisciplinary and Applied Sciences, Central University of Haryana, Mahendragarh, India.
| |
Collapse
|
20
|
Li M, Wu X, Guo Z, Gao R, Ni Z, Cui H, Zong M, Van Bockstaele F, Lou W. Lactiplantibacillus plantarum enables blood urate control in mice through degradation of nucleosides in gastrointestinal tract. MICROBIOME 2023; 11:153. [PMID: 37468996 PMCID: PMC10354915 DOI: 10.1186/s40168-023-01605-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/19/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Lactobacillus species in gut microbiota shows great promise in alleviation of metabolic diseases. However, little is known about the molecular mechanism of how Lactobacillus interacts with metabolites in circulation. Here, using high nucleoside intake to induce hyperuricemia in mice, we investigated the improvement in systemic urate metabolism by oral administration of L. plantarum via different host pathways. RESULTS Gene expression analysis demonstrated that L. plantarum inhibited the activity of xanthine oxidase and purine nucleoside phosphorylase in liver to suppress urate synthesis. The gut microbiota composition did not dramatically change by oral administration of L. plantarum over 14 days, indicated by no significant difference in α and β diversities. However, multi-omic network analysis revealed that increase of L. plantarum and decrease of L. johnsonii contributed to a decrease in serum urate levels. Besides, genomic analysis and recombinant protein expression showed that three ribonucleoside hydrolases, RihA-C, in L. plantarum rapidly and cooperatively catalyzed the hydrolysis of nucleosides into nucleobases. Furthermore, the absorption of nucleobase by intestinal epithelial cells was less than that of nucleoside, which resulted in a reduction of urate generation, evidenced by the phenomenon that mice fed with nucleobase diet generated less serum urate than those fed with nucleoside diet over a period of 9-day gavage. CONCLUSION Collectively, our work provides substantial evidence identifying the specific role of L. plantarum in improvement of urate circulation. We highlight the importance of the enzymes RihA-C existing in L. plantarum for the urate metabolism in hyperuricemia mice induced by a high-nucleoside diet. Although the direct connection between nucleobase transport and host urate levels has not been identified, the lack of nucleobase transporter in intestinal epithelial cells might be important to decrease its absorption and metabolization for urate production, leading to the decrease of serum urate in host. These findings provide important insights into urate metabolism regulation. Video Abstract.
Collapse
Affiliation(s)
- Mengfan Li
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Food Structure and Function Research Group (FSF), Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Xiaoling Wu
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Zewang Guo
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Ruichen Gao
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Zifu Ni
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Hualing Cui
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Minhua Zong
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Filip Van Bockstaele
- Food Structure and Function Research Group (FSF), Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.
| | - Wenyong Lou
- Lab of Applied Biocatalysis, School of Food Science and Engineering, South China University of Technology, Guangzhou, China.
| |
Collapse
|
21
|
Liu X, Ke L, Lei K, Yu Q, Zhang W, Li C, Tian Z. Antibiotic-induced gut microbiota dysbiosis has a functional impact on purine metabolism. BMC Microbiol 2023; 23:187. [PMID: 37442943 PMCID: PMC10339580 DOI: 10.1186/s12866-023-02932-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Dysbiosis of the gut microbiota is closely linked to hyperuricemia. However, the effect of the microbiome on uric acid (UA) metabolism remains unclear. This study aimed to explore the mechanisms through which microbiomes affect UA metabolism with the hypothesis that modifying the intestinal microbiota influences the development of hyperuricemia. RESULTS We proposed combining an antibiotic strategy with protein-protein interaction analysis to test this hypothesis. The data demonstrated that antibiotics altered the composition of gut microbiota as UA increased, and that the spectrum of the antibiotic was connected to the purine salvage pathway. The antibiotic-elevated UA concentration was dependent on the increase in microbiomes that code for the proteins involved in purine metabolism, and was paralleled by the depletion of bacteria-coding enzymes required for the purine salvage pathway. On the contrary, the microbiota with abundant purine salvage proteins decreased hyperuricemia. We also found that the antibiotic-increased microbiota coincided with a higher relative abundance of bacteria in hyperuricemia mice. CONCLUSIONS An antibiotic strategy combined with the prediction of microbiome bacterial function presents a feasible method for defining the key bacteria involved in hyperuricemia. Our investigations discovered that the core microbiomes of hyperuricemia may be related to the gut microbiota that enriches purine metabolism related-proteins. However, the bacteria that enrich the purine salvage-proteins may be a probiotic for decreasing urate, and are more likely to be killed by antibiotics. Therefore, the purine salvage pathway may be a potential target for the treatment of both hyperuricemia and antibiotic resistance.
Collapse
Affiliation(s)
- Xin Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China
| | - Leyong Ke
- Department of Cosmetic surgery, Kunming Medical University, Kunming, 650000, China
| | - Ke Lei
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Qian Yu
- Center of Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Wenqing Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China
| | - Changgui Li
- Institute of Metabolic Diseases, Qingdao University, Qingdao, 266003, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, China.
| |
Collapse
|
22
|
Lin X, Wang M, He Z, Hao G. Gut microbiota mediated the therapeutic efficiency of Simiao decoction in the treatment of gout arthritis mice. BMC Complement Med Ther 2023; 23:206. [PMID: 37344836 DOI: 10.1186/s12906-023-04042-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Gut microbiota plays a significant role in the development and treatment of gouty arthritis. Simiao decoction has been shown to alleviate gouty arthritis by inhibiting inflammation, regulating NLRP3 inflammasome, and altering gut microbiota. However, there is no evidence to prove whether gut microbiota directly mediates the therapeutic efficiency of Simiao decoction in treating gout arthritis. METHODS In this study, fecal microbiota transplantation (FMT) was used to transfer the gut microbiota of gout arthritis mice treated with Simiao decoction or allopurinol to blank gout arthritis mice, in order to investigate whether FMT had therapeutic effects on gout arthritis. RESULTS Both Simiao decoction and allopurinol effectively reduced the levels of serum uric acid, liver XOD activity, foot thickness, serum IL-1β, and G-CSF in gout arthritis mice. However, Simiao decoction also had additional benefits, including raising the pain threshold, reducing serum TNF-α and IL-6, alleviating gut inflammation, and repairing intestinal pathology, which were not observed with allopurinol treatment. Moreover, Simiao decoction had a greater impact on gut microbiota than allopurinol, as it was able to restore the abundance of phylum Proteobacteria and genus Helicobacter. After transplantation into gout arthritis mice, gut microbiota altered by Simiao decoction exhibited similar therapeutic effects to those of Simiao decoction, but gut microbiota altered by allopurinol showed no therapeutic effect. CONCLUSIONS These findings demonstrates that Simiao decoction can alleviate gout arthritis symptoms by regulating gut microbiota.
Collapse
Affiliation(s)
- Xiaoying Lin
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- Institute of Basic Research in Clinical Medicine, School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mingzhu Wang
- Institute of Basic Research in Clinical Medicine, School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhixing He
- Institute of Basic Research in Clinical Medicine, School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Guifeng Hao
- Institute of Basic Research in Clinical Medicine, School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, China.
| |
Collapse
|
23
|
Shirvani-Rad S, Khatibzade-Nasari N, Ejtahed HS, Larijani B. Exploring the role of gut microbiota dysbiosis in gout pathogenesis: a systematic review. Front Med (Lausanne) 2023; 10:1163778. [PMID: 37265486 PMCID: PMC10230090 DOI: 10.3389/fmed.2023.1163778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Objectives Gut dysbiosis is believed to be one of the several mechanisms that are involved in the pathogenesis of gout. This systematic review aimed to summarize the role of gut dysbiosis in gout disease and uncover the underlying mechanisms. Methods A comprehensive search was conducted on PubMed, Web of Science, and Scopus databases up to October 2021. Animal studies and human observational studies, including case-control, cross-sectional, and cohort studies assessing the association between gut microbiota composition and gout were included. The quality of included studies has been evaluated using the Newcastle-Ottawa Quality Assessment scale (NOS) and the SYRCLE's risk of bias tool. Results Initially, we found 274 studies among which 15 studies were included in this systematic review. Of them, 10 studies were conducted on humans and 5 studies were conducted on animals. Increased abundance of Alistipes and decreased abundance of Enterobacteriaceae alters purine metabolism, thereby aggravating gout condition. Moreover, a higher abundance of Phascolarctobacterium and Bacteroides in gout modulates enzymatic activity in purine metabolism. Butyrate-producing bacteria such as Faecalibacterium, prausnitzii, Oscillibacter, Butyricicoccus, and Bifidobacterium have higher abundance in healthy controls compared to gout patients, suggesting the anti-inflammatory and anti-microbial role of short-chain fatty acids (SCFAs). Lipopolysaccharides (LPS)-releasing bacteria, such as Enterobacteriaceae, Prevotella, and Bacteroides, are also involved in the pathogenesis of gout disease by stimulating the innate immune system. Conclusion Exploring the role of gut dysbiosis in gout and the underlying mechanisms can help develop microbiota-modulating therapies for gout.
Collapse
Affiliation(s)
- Salman Shirvani-Rad
- Microbiota Research Group, Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Faculty of Medicine, Qeshm International Medical Sciences Branch, Islamic Azad University, Qeshm, Iran
| | - Niloufar Khatibzade-Nasari
- Faculty of Medicine, Qeshm International Medical Sciences Branch, Islamic Azad University, Qeshm, Iran
- Young Researchers and Elite Club, Qeshm International Medical Sciences Branch, Islamic Azad University, Qeshm, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Hou T, Dai H, Wang Q, Hou Y, Zhang X, Lin H, Wang S, Li M, Zhao Z, Lu J, Xu Y, Chen Y, Gu Y, Zheng J, Wang T, Wang W, Bi Y, Ning G, Xu M. Dissecting the causal effect between gut microbiota, DHA, and urate metabolism: A large-scale bidirectional Mendelian randomization. Front Immunol 2023; 14:1148591. [PMID: 37063923 PMCID: PMC10097983 DOI: 10.3389/fimmu.2023.1148591] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/13/2023] [Indexed: 04/01/2023] Open
Abstract
ObjectivesOur aim was to investigate the interactive causal effects between gut microbiota and host urate metabolism and explore the underlying mechanism using genetic methods.MethodsWe extracted summary statistics from the abundance of 211 microbiota taxa from the MiBioGen (N =18,340), 205 microbiota metabolism pathways from the Dutch Microbiome Project (N =7738), gout from the Global Biobank Meta-analysis Initiative (N =1,448,128), urate from CKDGen (N =288,649), and replication datasets from the Global Urate Genetics Consortium (N gout =69,374; N urate =110,347). We used linkage disequilibrium score regression and bidirectional Mendelian randomization (MR) to detect genetic causality between microbiota and gout/urate. Mediation MR and colocalization were performed to investigate potential mediators in the association between microbiota and urate metabolism.ResultsTwo taxa had a common causal effect on both gout and urate, whereas the Victivallaceae family was replicable. Six taxa were commonly affected by both gout and urate, whereas the Ruminococcus gnavus group genus was replicable. Genetic correlation supported significant results in MR. Two microbiota metabolic pathways were commonly affected by gout and urate. Mediation analysis indicated that the Bifidobacteriales order and Bifidobacteriaceae family had protective effects on urate mediated by increasing docosahexaenoic acid. These two bacteria shared a common causal variant rs182549 with both docosahexaenoic acid and urate, which was located within MCM6/LCT locus.ConclusionsGut microbiota and host urate metabolism had a bidirectional causal association, implicating the critical role of host-microbiota crosstalk in hyperuricemic patients. Changes in gut microbiota can not only ameliorate host urate metabolism but also become a foreboding indicator of urate metabolic diseases.
Collapse
Affiliation(s)
- Tianzhichao Hou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huajie Dai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanan Hou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyun Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyun Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Min Xu,
| |
Collapse
|
25
|
Fernández-Torres J, Zamudio-Cuevas Y, Martínez-Nava GA, Martínez-Flores K, Ruíz-Dávila X, Sánchez-Sánchez R. Relationship between rs4349859 and rs116488202 polymorphisms close to MHC-I region and serum urate levels in patients with gout. Mol Biol Rep 2023; 50:4367-4374. [PMID: 36943604 DOI: 10.1007/s11033-023-08359-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/23/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Gout is the most common inflammatory rheumatic disease and elevated levels of serum urate (SU) are the main cause for its development. Major histocompatibility complex class 1 (MHC-1) plays an important role in the development of multiple inflammatory diseases; however, there is little evidence of its involvement in gout. The present study focused on evaluating the association of the rs4349859 and rs116488202 single nucleotide polymorphisms (SNPs) close to the MHC-1 region in patients with gout. METHODS AND RESULTS One hundred and seventy-six individuals of Mexican origin were included, of which 81 were patients with primary gout and 95 were healthy controls. The rs4349859 and rs116488202 SNPs were genotyped using TaqMan probes by allelic discrimination by real-time PCR. Serum concentrations of biochemical parameters were measured with enzymatic methods. Descriptive statistics were applied and P-values < 0.05 were considered significant. It was observed that the rs4349859 and rs116488202 SNPs showed significant association with the risk of gout (OR = 146, 95%CI = 44.8-480.2, P < 0.01; OR = 2885, 95%CI = 265-31398, P < 0.01, respectively). Our results also showed significantly higher serum SU levels in gout patients with respect to controls (P < 0.01) in the carriers of the GA genotype compared with the GG genotype of the rs4349859 variant, and in the carriers of the CT genotype compared with the CC genotype of the rs116488202 variant. CONCLUSION The study revealed that rs4349859 and rs116488202 SNPs close to MHC-I region confers strong susceptibility to gout in Mexican population, and the heterozygous genotypes of both were associated with higher levels of SU.
Collapse
Affiliation(s)
- Javier Fernández-Torres
- Synovial Fluid Laboratory, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico.
- Biology Department, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.
| | - Yessica Zamudio-Cuevas
- Synovial Fluid Laboratory, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | | | - Karina Martínez-Flores
- Synovial Fluid Laboratory, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | | | - Roberto Sánchez-Sánchez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| |
Collapse
|
26
|
The impact of short-chain fatty acid-producing bacteria of the gut microbiota in hyperuricemia and gout diagnosis. Clin Rheumatol 2023; 42:203-214. [PMID: 36201123 DOI: 10.1007/s10067-022-06392-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 01/11/2023]
Abstract
INTRODUCTION/OBJECTIVES Persistent hyperuricemia is a key factor in gout; however, only 13.5% of hyperuricemic individuals manifest the disease. The gut microbiota could be one of the many factors underlying this phenomenon. We aimed to assess the difference in taxonomic and predicted functional profiles of the gut microbiota between asymptomatic hyperuricemia (AH) individuals and gout patients. METHODS The V3-V4 region of the 16S rRNA gene of the gut microbiota of AH individuals, gout patients, and controls was sequenced. Bioinformatic analyses were carried out with QIIME2 and phyloseq to determine the difference in the relative abundance of bacterial genera among the study groups. Tax4fun2 was used to predict the functional profile of the gut microbiota. RESULTS AH individuals presented a higher abundance of butyrate- and propionate-producing bacteria than gout patients; however, the latter had more bacteria capable of producing acetate. The abundance of Prevotella genus bacteria was not significantly different between the patients but was higher than that in controls. This result was corroborated by the functional profile, in which AH individuals had less pyruvate oxidase abundance than gout patients and less abundance of an enzyme that regulates glutamate synthetase activation than controls. CONCLUSION We observed a distinctive taxonomic profile in AH individuals characterized by a higher abundance of short-chain fatty acid-producing bacteria in comparison to those observed in gout patients. Furthermore, we provide scientific evidence that indicates that the gut microbiota of AH individuals could provide anti-inflammatory mediators, which prevent the appearance of gout flares. Key Points • AH and gout patients both have a higher abundance of Prevotella genus bacteria than controls. • AH individuals' gut microbiota had more butyrate- and propionate-producing bacteria than gout patients. • The gut microbiome of AH individuals provides anti-inflammatory mediators that could prevent gout flares.
Collapse
|
27
|
Chen Z, Wang Z, Li D, Zhu B, Xia Y, Wang G, Ai L, Zhang C, Wang C. The gut microbiota as a target to improve health conditions in a confined environment. Front Microbiol 2022; 13:1067756. [PMID: 36601399 PMCID: PMC9806127 DOI: 10.3389/fmicb.2022.1067756] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
Confined environments increase psychological stress and lead to health problems such as abnormal mood and rhythm disruption. However, the mechanism by which confined environments impact health has remained unclear. Significant correlations have been reported between psychological stress and changes in gut microbiota. Therefore, we investigated the effect of a confined environment on the composition of the gut microbiota by 16s rDNA high-throughput sequencing, and analyzed the correlation between gut microbiota and health indicators such as uric acid (UA), sleep, and mood. We found that the gut microbiota of the subjects clustered into two enterotypes (Bi and Bla), and that the groups differed significantly. There were notable differences in the abundances of genera such as Bifidobacterium, Dorea, Ruminococcus_torques_group, Ruminococcus_gnavus_group, Klebsiella, and UCG-002 (p < 0.05). A confined environment significantly impacted the subjects' health indicators. We also observed differences in how the subjects of the two enterotypes adapted to the confined environment. The Bi group showed no significant differences in health indicators before and after confinement; however, the Bla group experienced several health problems after confinement, such as increased UA, anxiety, and constipation, and lack of sleep. Redundancy analysis (RDA) showed that UA, RBC, mood, and other health problems were significantly correlated with the structure of the gut microbiota. We concluded that genera such as UCG-002, Ruminococcus, CAG352, and Ruminococcus_torques_group increased vulnerability to confined environments, resulting in abnormal health conditions. We found that the differences in the adaptability of individuals to confined environments were closely related to the composition of their gut microbiota.
Collapse
Affiliation(s)
- Zheng Chen
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - ZiYing Wang
- Navy Special Medical Center, Naval Medical University, Shanghai, China
| | - Dan Li
- Navy Special Medical Center, Naval Medical University, Shanghai, China
| | - Beiwei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Yongjun Xia
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Guangqiang Wang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Lianzhong Ai
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Chunhong Zhang
- Navy Special Medical Center, Naval Medical University, Shanghai, China,*Correspondence: Chunhong Zhang,
| | - Chuan Wang
- Navy Special Medical Center, Naval Medical University, Shanghai, China,Chuan Wang,
| |
Collapse
|
28
|
Wang X, Long H, Chen M, Zhou Z, Wu Q, Xu S, Li G, Lu Z. Modified Baihu decoction therapeutically remodels gut microbiota to inhibit acute gouty arthritis. Front Physiol 2022; 13:1023453. [PMID: 36589463 PMCID: PMC9798006 DOI: 10.3389/fphys.2022.1023453] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Acute gouty arthritis (AGA) is the most common first symptom of gout, and the development of gout as a metabolic and immune inflammatory disease is also correlated with the gut microbiota. However, the mechanism of the effect of changes in the gut microbiota on AGA remains unclear. The intestinal flora can not only affect purine metabolism or regulate inflammation, but also influence the therapeutic effect of drugs on AGA. The aim of this study was to investigate the exact mechanism of modified Baihu decoction (MBD) in the treatment of AGA and whether it is related to the regulation of the structure of the intestinal flora. Methods: On the 21st day of MBD administration by continuous gavage, a rat acute gouty arthritis model was constructed using sodium urate (0.1 mL/rat, 50 mg/mL), and the ankle joint swelling was measured before and 4 h, 8 h, 24 h, and 48 h after the injection of sodium urate. After 48 h of sodium urate injection, serum, liver, kidney, ankle synovial tissue and feces were collected from rats. The collected samples were examined and analyzed using H&E, Elisa, Immunohistochemistry, Histopathology, 16S rDNA, and Biochemical analysis. To investigate the mechanism of MBD to alleviate AGA using pro-inflammatory factors and intestinal flora. Results: MBD (5.84, 35 g/kg) was administered orally to AGA rats and diclofenac sodium tablets (DS-tablets) were used as standard treatment control. Serum biochemical assessment confirmed that MBD is a safe drug for the treatment of AGA. In addition, our findings confirmed that MBD relieved AGA-related symptoms, such as toe swelling. Lowering serum levels of uric acid, IL-1β, and TGF-β1 immunohistochemical results also confirmed that MBD reduced the expression of inflammatory elements such as IL-1β, NLRP3, ASC, and Caspase-1 in synovial tissue.Furthermore, compared with control group, the 16s rDNA sequencing of AGA rat faeces revealed an increase in the relative abundance of Lachnospiraceae, Muribaculaceae, and Bifidobacteriaceae species. While the relative abundance of Lactobacillaceae, Erysipelotrichaceae, Ruminococcaceae, Prevotellaceae and Enterobacteriaceae showed a relative decrease in species abundance. Of these, the reduction in species abundance of Enterobacteriaceae was associated with a reduction in amino acid metabolism and environmental perception. After MBD therapeutic intervention, the disturbance of the intestinal flora caused by AGA was restored. Conclusion: In summary, MBD is an effective agent for the treatment of AGA, with the potential mechanism being the regulation of intestinal flora to control inflammation. This would help to promote the therapeutic effect of MBD on AGA.
Collapse
Affiliation(s)
- Xianyang Wang
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haishan Long
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming Chen
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China
| | - Zongbo Zhou
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China
| | - Qinlin Wu
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China
| | - Shijie Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Shijie Xu, ; Geng Li, ; Zhifu Lu,
| | - Geng Li
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Shijie Xu, ; Geng Li, ; Zhifu Lu,
| | - Zhifu Lu
- Haikou Hospital of Traditional Chinese Medicine, Haikou, Hainan, China,*Correspondence: Shijie Xu, ; Geng Li, ; Zhifu Lu,
| |
Collapse
|
29
|
Tong S, Zhang P, Cheng Q, Chen M, Chen X, Wang Z, Lu X, Wu H. The role of gut microbiota in gout: Is gut microbiota a potential target for gout treatment. Front Cell Infect Microbiol 2022; 12:1051682. [PMID: 36506033 PMCID: PMC9730829 DOI: 10.3389/fcimb.2022.1051682] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
Numerous studies have demonstrated that gut microbiota is essential for the host's health because it regulates the host's metabolism, endocrine, and immune systems. In recent years, increasing evidence has shown that gut microbiota plays a role in the onset and progression of gout. Changes in the composition and metabolism of the gut microbiota, result in abnormalities of uric acid degradation, increasing uric acid generation, releasing pro-inflammatory mediators, and intestinal barrier damage in developing gout. As a result, gout therapy that targets gut microbiota has drawn significant interest. This review summarized how the gut microbiota contributes to the pathophysiology of gout and how gout affects the gut microbiota. Additionally, this study explained how gut microbiota might serve as a unique index for the diagnosis of gout and how conventional gout treatment medicines interact with it. Finally, prospective therapeutic approaches focusing on gut microbiota for the prevention and treatment of gout were highlighted, which may represent a future avenue in gout treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaoyong Lu
- *Correspondence: Xiaoyong Lu, ; Huaxiang Wu,
| | - Huaxiang Wu
- *Correspondence: Xiaoyong Lu, ; Huaxiang Wu,
| |
Collapse
|
30
|
Metagenomic Analysis of Gut Microbiome in Gout Patients with Different Chinese Traditional Medicine Treatments. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6466149. [PMID: 36262169 PMCID: PMC9576389 DOI: 10.1155/2022/6466149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/30/2022] [Indexed: 11/21/2022]
Abstract
Introduction Changes in eating habits have made gout a metabolic disease of increasing concern. Previous studies have indicated that there are significant differences in species composition and abundance of gut microbiome in gout patients compared with average. Considering that traditional Chinese medicine has a momentous effect in treating gout, the research study aimed to explore the differences of genomic and metabolomics of gut microbiome before and after traditional Chinese medicine treatment in patients with gout. Method 30 patients with gout and 29 matched controls were recruited of which 16 patients took H treatment and 14 patients took T treatment. Stools were collected twice for patients before and after treatment and only once for controls. A total of 89 samples were annotated with metagenomic species and functions, and the enrichment analysis of differential genes and KO pathway was carried out. Result The results showed a decrease in the diversity of gut microbiome in gout patients and the gene abundance and metabolomics had great differences among study groups. The number of bacterial genera also had significant differences among treatment groups. Moreover, among different groups, the regulation of different species was variously correlated. The correlation between species and clinical laboratory indicators in the rising group was stronger than that in the decreasing group and the upregulation of some strain was related to the content of urea nitrogen. Conclusion After the traditional Chinese medicine treatment, the glutathione pathway was significantly enriched and some pathogenic bacteria were significantly inhibited. The study suggests that traditional Chinese medicine treatment may exert its therapeutic effect by inhibiting relevant pathways.
Collapse
|
31
|
Li Y, Zhu J, Lin G, Gao K, Yu Y, Chen S, Chen L, Chen Z, Li L. Probiotic effects of Lacticaseibacillus rhamnosus 1155 and Limosilactobacillus fermentum 2644 on hyperuricemic rats. Front Nutr 2022; 9:993951. [PMID: 36245501 PMCID: PMC9562091 DOI: 10.3389/fnut.2022.993951] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Hyperuricemia is the main cause of gout and involved in the occurrence of multiple diseases, such as hypertension, metabolic disorders and chronic kidney disease. Emerging evidence suggests that lactic acid bacteria (LAB) have shown the beneficial effects on the prevention or treatment of hyperuricemia. In this study, the urate-lowering effect of two LAB strains, Lacticaseibacillus rhamnosus 1155 (LR1155) and Limosilactobacillus fermentum 2644 (LF2644) on hyperuricemic rats were investigated. A hyperuricemic rat model was induced by the intragastric treatment of potassium oxonate, combined with a high purine diet. The oral administration of LR1155, LF2644, or a combination of LR1155 and LF2644 for 4 weeks significantly prevented the rise of the serum uric acid (UA) induced by hyperuricemia. LR1155 and LF2644 significantly elevated the fecal UA levels, increased the UA content and up-regulated gene expression of UA transporter, ATP-binding cassette subfamily G-2 (ABCG2), in colon and jejunum tissues, suggesting the accelerated UA excretion from the intestine. Besides, LR1155 significantly inhibited the activity of xanthine oxidase (XOD) in liver and serum, benefited the reduce of UA production. In addition, LF2644 strengthened the gut barrier functions through an up-regulation of the gene expressions for occluding and mucin2, accompanied with the reduced inflammatory indicators of lipopolysaccharide (LPS) and interleukin-1β (IL-1β) in hyperuricemic rat. Moreover, using 16s rDNA high-throughput sequencing of feces, LR1155 was shown to improve the hyperuricemia induced gut microbial dysbiosis. The genera Roseburia, Butyricicoccus, Prevotella, Oscillibacter, and Bifidobacterium may associate with the effect of LR1155 on microbiota in hyperuricemic rats. Collectively, the results indicated that LR1155 and LF2644 exhibit urate-lowering effects and could be used alone or in combination as a new adjuvant treatment for hyperuricemia.
Collapse
Affiliation(s)
- Yanjun Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Jun Zhu
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- *Correspondence: Jun Zhu,
| | - Guodong Lin
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Kan Gao
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Yunxia Yu
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Su Chen
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Lie Chen
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Zuoguo Chen
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| | - Li Li
- Department of Research and Development, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
| |
Collapse
|
32
|
Sun L, Ni C, Zhao J, Wang G, Chen W. Probiotics, bioactive compounds and dietary patterns for the effective management of hyperuricemia: a review. Crit Rev Food Sci Nutr 2022; 64:2016-2031. [PMID: 36073759 DOI: 10.1080/10408398.2022.2119934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Hyperuricemia is closely linked with an increased risk of developing hypertension, diabetes, renal failure and other metabolic syndromes. Probiotics, bioactive compounds and dietary patterns are safe cost-efficient ways to control hyperuricemia, whereas comprehensive reviews of their anti-hyperuricemic mechanisms are limited. This review summarizes the roles of probiotics, bioactive compounds and dietary patterns in treating hyperuricemia and critically reviews the possible mechanisms by which these interventions exert their activities. The dietary patterns are closely related to the occurrence of hyperuricemia through the indirect action of gut microbiota or the direct effects of host purine metabolism. The Mediterranean and Dietary Approaches to Stop Hypertension diets help reduce serum uric acid concentrations and thus prevent hyperuricemia. Meanwhile, probiotics alleviate hyperuricemia by ways of absorbing purine, restoring gut microbiota dysbiosis and inhibiting xanthine oxidase (XO) activity. Bioactive compounds such as polyphenols, peptides and alkaloids exert various anti-hyperuricemic effects, by regulating urate transporters, blocking the active sites of XO and inhibiting the toll-like receptor 4/nuclear factor kappa B signaling pathway and NOD-, LRR- and pyrin domain-containing protein 3 signaling pathway. This review will assist people with hyperuricemia to adopt a healthy diet and contribute to the application of natural products with anti-hyperuricemic activity.
Collapse
Affiliation(s)
- Lei Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Caixin Ni
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
33
|
Palacios-González B, León-Reyes G, Rivera-Paredez B, Ibarra-González I, Vela-Amieva M, Flores YN, Canizales-Quinteros S, Salmerón J, Velázquez-Cruz R. Targeted Metabolomics Revealed a Sex-Dependent Signature for Metabolic Syndrome in the Mexican Population. Nutrients 2022; 14:nu14183678. [PMID: 36145054 PMCID: PMC9504093 DOI: 10.3390/nu14183678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
Metabolic syndrome (MetS) is a group of several metabolic conditions predisposing to chronic diseases. Individuals diagnosed with MetS are physiologically heterogeneous, with significant sex-specific differences. Therefore, we aimed to investigate the potential sex-specific serum modifications of amino acids and acylcarnitines (ACs) and their relationship with MetS in the Mexican population. This study included 602 participants from the Health Workers Cohort Study. Forty serum metabolites were analyzed using a targeted metabolomics approach. Multivariate regression models were used to test associations of clinical and biochemical parameters with metabolomic profiles. Our findings showed a serum amino acid signature (citrulline and glycine) and medium-chain ACs (AC14:1, AC10, and AC18:10H) associated with MetS. Glycine and AC10 were specific metabolites representative of discrimination according to sex-dependent MetS. In addition, we found that glycine and short-chain ACs (AC2, AC3, and AC8:1) are associated with age-dependent MetS. We also reported a significant correlation between body fat and metabolites associated with sex-age-dependent MetS. In conclusion, the metabolic profile varies by MetS status, and these differences are sex-age-dependent in the Mexican population.
Collapse
Affiliation(s)
| | - Guadalupe León-Reyes
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
| | - Berenice Rivera-Paredez
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | | | - Marcela Vela-Amieva
- Laboratory of Inborn Errors of Metabolism, National Pediatrics Institute (INP), Mexico City 04530, Mexico
| | - Yvonne N. Flores
- Epidemiological and Health Services Research Unit, Morelos Mexican Institute of Social Security, Cuernavaca 62000, Mexico
- Department of Health Policy and Management and UCLA-Kaiser Permanente Center for Health Equity, Fielding School of Public Health, University of California, Los Angeles, CA 90095, USA
- UCLA Center for Cancer Prevention and Control Research, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Samuel Canizales-Quinteros
- Unit of Genomics of Population Applied to Health, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
- National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
| | - Jorge Salmerón
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Rafael Velázquez-Cruz
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
- Correspondence: ; Tel./Fax: +52-(55)-5350-1900
| |
Collapse
|
34
|
Fernández-Torres J, Martínez-Nava GA, Martínez-Flores K, Sánchez-Sánchez R, Jara LJ, Zamudio-Cuevas Y. The interplay between HLA-B and NLRP3 polymorphisms may be associated with the genetic susceptibility of gout. Mol Biol Rep 2022; 49:10205-10215. [PMID: 36057006 DOI: 10.1007/s11033-022-07895-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND HLA and NLRP3 play an important role in the development of various autoimmune and autoinflammatory diseases. Gout is an autoinflammatory disease associated with multiple genetic and environmental factors. The objective of the present study was to evaluate the interaction and association between genetic polymorphisms of HLA-B and the NLRP3 gene in Mexican patients with gout. METHODS AND RESULTS Eighty-one patients with gout were included and compared with 95 healthy subjects. The polymorphisms rs4349859, rs116488202, rs2734583 and rs3099844 (within the HLA-B region) and rs3806268 and rs10754558 of the NLRP3 gene were genotyped using TaqMan probes in a Rotor-Gene device. The interactions were determined using the multifactorial dimensionality reduction (MDR) method, while the associations were determined through logistic regression models. The MDR analysis revealed significant interactions between the rs116488202 and rs10754558 polymorphisms with an entropy value of 4.31% (p < 0.0001). Significant risk associations were observed with rs4349859 and rs116488202 polymorphisms (p < 0.01); however, no significant associations were observed with the polymorphisms of the NLRP3 gene. CONCLUSIONS The results suggest that HLA-B polymorphisms and their interaction with NLRP3 may contribute to the genetic susceptibility of gout.
Collapse
Affiliation(s)
- Javier Fernández-Torres
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico.,Biology Department, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | | | - Karina Martínez-Flores
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Roberto Sánchez-Sánchez
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Luis J Jara
- Rheumatology Division, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico
| | - Yessica Zamudio-Cuevas
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Mexico City, Mexico.
| |
Collapse
|
35
|
Wang Z, Li Y, Liao W, Huang J, Liu Y, Li Z, Tang J. Gut microbiota remodeling: A promising therapeutic strategy to confront hyperuricemia and gout. Front Cell Infect Microbiol 2022; 12:935723. [PMID: 36034697 PMCID: PMC9399429 DOI: 10.3389/fcimb.2022.935723] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
The incidence of hyperuricemia (HUA) and gout continuously increases and has become a major public health problem. The gut microbiota, which colonizes the human intestine, has a mutually beneficial and symbiotic relationship with the host and plays a vital role in the host's metabolism and immune regulation. Structural changes or imbalance in the gut microbiota could cause metabolic disorders and participate in the synthesis of purine-metabolizing enzymes and the release of inflammatory cytokines, which is closely related to the occurrence and development of the metabolic immune disease HUA and gout. The gut microbiota as an entry point to explore the pathogenesis of HUA and gout has become a new research hotspot. This review summarizes the characteristics of the gut microbiota in patients with HUA and gout. Meanwhile, the influence of different dietary structures on the gut microbiota, the effect of the gut microbiota on purine and uric acid metabolism, and the internal relationship between the gut microbiota and metabolic endotoxemia/inflammatory factors are explored. Moreover, the intervention effects of probiotics, prebiotics, and fecal microbial transplantation on HUA and gout are also systematically reviewed to provide a gut flora solution for the prevention and treatment of related diseases.
Collapse
Affiliation(s)
- Zhilei Wang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuchen Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenhao Liao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ju Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanping Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhiyong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
36
|
Yuan X, Chen R, Zhang Y, Lin X, Yang X. Altered Gut Microbiota in Children With Hyperuricemia. Front Endocrinol (Lausanne) 2022; 13:848715. [PMID: 35574004 PMCID: PMC9091909 DOI: 10.3389/fendo.2022.848715] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
Background In adults, gut dysbiosis may contribute to the pathogenesis of gout. However, the characteristics of gut microbiota in children with hyperuricemia (HUA) in the absence of clinical gout have not been explored. Objective This present study analyzed the gut microbiota in children with HUA as compared to controls (Con) and explored bacterial associations that may account for differences. Methods A total of 80 children were enrolled in this study; they were divided into HUA and Con according to the level of serum uric acid (UA). The composition of gut microbiota was investigated by 16S rRNA high-throughput sequencing. Results Principal coordinate analysis revealed that gut microbiota of the HUA group was clustered together and separated partly from the Con group. There was no difference in alpha-diversity between the two groups. However, Spearman's correlation analysis revealed that serum UA level positively correlated with genera Actinomyces, Morganella, and Streptococcus, and negatively associated with the producers of short-chain fatty acids (SCFAs), such as Alistipes, Faecalibacterium, and Oscillospira, and the sulfidogenic bacteria Bilophila. The members of the genera Alistipes and Bilophila in the Con group were significantly more prevalent than the HUA subjects. Compared to the Con cohort, metabolic pathway predictions found that the superpathways of purine nucleotide de novo biosynthesis were decreased in HUA subjects, whereas the superpathway of purine deoxyribonucleoside de gradation was increased. Conclusion The composition of the gut microbiota in children with HUA differs from Con. Although causality cannot be established, modification in the microbiota that produces SCFA and sulfide may promote HUA.
Collapse
Affiliation(s)
| | - Ruimin Chen
- Department of Endocrinology, Genetics and Metabolism, Fuzhou Children’s Hospital of Fujian Medical University, Fuzhou, China
| | | | | | | |
Collapse
|
37
|
H19 is involved in the regulation of inflammatory responses in acute gouty arthritis by targeting miR-2-3p. Immunol Res 2022; 70:392-399. [PMID: 35314952 DOI: 10.1007/s12026-022-09276-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/10/2022] [Indexed: 11/05/2022]
Abstract
A great number of studies have confirmed that long noncoding RNA (lncRNA) are involved in the regulation of inflammatory response in acute gouty arthritis (AGA). This paper aimed to survey the regulatory mechanism of H19 on AGA. The expression of serum H19 in all subjects was examined by qRT-PCR. The ROC curve was used to estimate the diagnostic value of H19 for AGA. THP-1 cells were induced by MSU to establish in vitro AGA cell model. The concentrations of cytokines such as IL-1β, IL-8, and TNF-α were tested by ELISA. Luciferase reporter gene analysis was used to verify the interaction between H19 and the 3'-UTR of miR-22-3p. Expressions of serum H19 in AGA patients were significantly higher than that in controls. The ROC curve indicated the potential of H19 as a diagnostic marker for AGA. Cell experiments revealed that the downregulation of H19 significantly inhibited the expressions of IL-1β, IL-8, and TNF-α. The luciferase reporter gene assay manifested that miR-22-3p is the target gene of H19. And knockdown of miR-22-3p overturned the downregulation of inflammatory factors caused by H19 inhibition. H19 aggravated MSU-induced THP-1 inflammation by negatively targeting miR-22-3p, suggesting a new regulatory mechanism and potential therapeutic target for AGA.
Collapse
|
38
|
Kim HW, Yoon EJ, Jeong SH, Park MC. Distinct Gut Microbiota in Patients with Asymptomatic Hyperuricemia: A Potential Protector against Gout Development. Yonsei Med J 2022; 63:241-251. [PMID: 35184426 PMCID: PMC8860935 DOI: 10.3349/ymj.2022.63.3.241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Here, we aimed to elucidate the differences in microbiota composition between patients with gout and those with asymptomatic hyperuricemia (asHU) and determine the effect of uric acid-lowering therapy (ULT) on the gut microbiome. MATERIALS AND METHODS Stool samples from patients with asHU (n=8) and three groups of gout patients, i.e., acute gout patients before ULT (0ULT, n=14), the same acute gout patients after 30-day ULT (30ULT, n=9), and chronic gout patients after ≥6-month ULT (cULT, n=18) were collected and analyzed using 16S rRNA gene-based pyrosequencing. The composition of microbial taxonomy and communities, species diversity, and relationships among microbial communities were elucidated by bioinformatic analysis. RESULTS Gout patients showed less diverse gut microbiota than asHU patients. The microbiota of the asHU group exhibited a higher Firmicutes-to-Bacteroidetes (F/B) ratio and lower Prevotella-to-Bacteroides (P/B) ratio than the gout group; significantly, the F/B ratio increased in gout patients after ULT. Moreover, a balanced enterotype populated asHU patients compared to gout patients. Notably, the gut microbiota in asHU patients had a higher proportion of taxa with potentially anti-inflammatory effects compared to the gut microbiota in gout patients. CONCLUSION We found that microbial composition differs between asHU and gout patients. The differential gut microbiota in asHU patients may protect against gout development, whereas that in gout patients may have a role in gout provocation. ULT in gout patients altered the gut microbiota, and may help alleviate gout pathology and mitigate gout progression.
Collapse
Affiliation(s)
- Hye Won Kim
- Department of Medicine, The Graduate School, Yonsei University College of Medicine, Seoul, Korea
- Hospital Medicine Center, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Eun-Jeong Yoon
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
- Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, Korea
| | - Seok Hoon Jeong
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
- Research Institute of Bacterial Resistance, Yonsei University College of Medicine, Seoul, Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
39
|
Park HK, Lee SJ. Treatment of gouty arthritis is associated with restoring the gut microbiota and promoting the production of short-chain fatty acids. Arthritis Res Ther 2022; 24:51. [PMID: 35183234 PMCID: PMC8857835 DOI: 10.1186/s13075-022-02742-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Although factors initiating the inflammatory response to monosodium urate crystals have been identified, the role of the gut microbiota and their metabolites on gout remains unknown. This study aimed to investigate the changes in both gut microbiota and short-chain fatty acids (SCFAs) according to inflammatory states of gout in the same patients. Methods This study enrolled 20 patients with gout in the acute state who had active joints and were followed up until the recovery state with no active joints. Blood and fecal samples were simultaneously collected within 3 days for each disease state. The stool microbiome was analyzed using 16S rRNA sequencing, and serum SCFAs were measured by gas chromatography-mass spectrometry. Differences in the gut microbiome and serum SCFAs were compared between the acute and recovery states. Results Beta diversity of the microbiome was significantly different between the acute and recovery states in terms of weighted UniFrac distance. In the recovery state, Prevotellaceae (p = 0.006) and the genus Prevotella (p = 0.009) were significantly enriched, whereas Enterobacteriaceae (p = 0.019) and its derivative genus Shigella (p = 0.023) were significantly decreased compared to the acute state. Similarly, the levels of acetate were dramatically increased in the recovery state compared to the acute state (p < 0.010). The levels of propionate and butyrate tended to increase but without statistical significance. Conclusion Substantial alterations of bacterial composition with the promotion of SCFA formation (especially acetate) were found after treatment in patients with gouty arthritis. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02742-9.
Collapse
|
40
|
Zhang W, Wang T, Guo R, Cui W, Yu W, Wang Z, Jiang Y, Jiang M, Wang X, Liu C, Xiao J, Shang J, Wen X, Zhao Z. Variation of Serum Uric Acid Is Associated With Gut Microbiota in Patients With Diabetes Mellitus. Front Cell Infect Microbiol 2022; 11:761757. [PMID: 35118005 PMCID: PMC8803748 DOI: 10.3389/fcimb.2021.761757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/15/2021] [Indexed: 02/04/2023] Open
Abstract
Diabetes mellitus is a metabolic disease closely related to a disordered gut microbiome. Diabetic patients usually suffer from various metabolic disorders, such as increased serum uric acid levels. Although serum uric acid levels depend partially on intestine excretion, the relationship between uric acid and gut microbiome in diabetic patients remains unknown. We collected a total of 126 fecal samples from diabetic patients for 16S ribosomal RNA gene amplicon sequencing and recorded clinical data. We analyzed the correlation between clinical indicators and gut microbiota of diabetic patients using Spearman analysis. Since uric acid was the most prominent one, we classified diabetic patients based on their uric acid levels to find the microbiome associated with uric acid disturbance. We constructed Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway profiles using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) to identify variations between the different groups. Among all the clinical indicators, uric acid had the strongest correlation with gut microbiota. First, we divided the patients into three groups according to their uric acid levels. The two low uric acid groups were similar, while the elevated uric acid group had significant differences in gut microbiota and metabolic pathways. The elevated uric acid group had a significantly lower gut microbiota diversity. At the genus level, this group had remarkably higher Escherichia–Shigella amounts and notably lower Faecalibacterium, Oscillospiraceae_UCG−002, and Oscillospiraceae_UCG−005 amounts. The gut microbiota of the high uric acid group was predicted to be enriched in metabolism, human diseases, and lipopolysaccharide biosynthesis. Since the two low uric acid groups were similar, we regrouped and matched the abnormal uric acid patients with normal uric acid patients. The differences in gut microbiota and metabolic pathways related to nucleotide metabolism became more significant. The serum uric acid levels were associated with gut microbiome changes. This might be related to uric acid metabolism by gut microbes. Our study indicates that targeting the gut microbiome could help manage elevated uric acid levels.
Collapse
Affiliation(s)
- Weifeng Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ting Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruixue Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wen Cui
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Yu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihui Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yumin Jiang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Minghan Jiang
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaojie Wang
- Department of Pharmacology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Chao Liu
- Shanghai Mobio Biomedical Technology Co., Ltd., Shanghai, China
| | - Jing Xiao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhanzheng Zhao, ; Xuejun Wen, ; Jin Shang,
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, United States
- *Correspondence: Zhanzheng Zhao, ; Xuejun Wen, ; Jin Shang,
| | - Zhanzheng Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhanzheng Zhao, ; Xuejun Wen, ; Jin Shang,
| |
Collapse
|
41
|
Yang HT, Xiu WJ, Liu JK, Yang Y, Hou XG, Zheng YY, Wu TT, Wu CX, Xie X. Gut Microbiota Characterization in Patients with Asymptomatic Hyperuricemia: probiotics increased. Bioengineered 2021; 12:7263-7275. [PMID: 34590550 PMCID: PMC8806635 DOI: 10.1080/21655979.2021.1976897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/31/2021] [Indexed: 10/27/2022] Open
Abstract
Asymptomatic hyperuricemia (AH) is an early stage of gout. Emerging evidence shows that the intestinal microbiota is related to gout. However, the relationship between AH and the intestinal microbiota is poorly understood. Therefore, the aim of the current study was to explore the possible correlation between AH and intestinal flora. We compared the intestinal microbial communities of AH (45 cases) and healthy subjects (45 cases) by 16S rRNA gene sequencing and clustering analysis on the incorporated population. Intestinal-type clustering can be divided into two groups, and significant differences in the proportion of AH are found among different bowel types. Alpha diversity indices were higher in the AH group than in the control group, and beta diversity indices also showed significant differences. A total of 19 genera were found different between the AH group and the control group. Compared with the control group, some probiotics are increased in the AH population. Two groups were ranked by importance of bacteria. We found the different bacteria partially coincided with the important bacteria, and the joint diagnosis level of the important bacteria was good. Conclusion: There were significant differences in the composition of intestinal biota between AH patients and healthy subjects. Some probiotics increased in AH.
Collapse
Affiliation(s)
- Hai-Tao Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wen-Juan Xiu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jing-Kun Liu
- Department of Oncology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yi Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chen-Xin Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
42
|
Méndez-Salazar EO, Martínez-Nava GA. Uric acid extrarenal excretion: the gut microbiome as an evident yet understated factor in gout development. Rheumatol Int 2021; 42:403-412. [PMID: 34586473 DOI: 10.1007/s00296-021-05007-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/19/2021] [Indexed: 12/19/2022]
Abstract
Humans do not produce uricase, an enzyme responsible for degrading uric acid. However, some bacteria residing in the gut can degrade one-third of the dietary and endogenous uric acid generated daily. New insights based on metagenomic and metabolomic approaches provide a new interest in exploring the involvement of gut microbiota in gout. Nevertheless, the exact mechanisms underlying this association are complex and have not been widely discussed. In this study, we aimed to review the evidence that suggests uric acid extrarenal excretion and gut microbiome are potential risk factors for developing gout. A literature search was performed in PubMed, Web of Science, and Google Scholar using several keywords, including "gut microbiome AND gout". A remarkable intestinal dysbiosis and shifts in abundance of certain bacterial taxa in gout patients have been consistently reported among different studies. Under this condition, bacteria might have developed adaptive mechanisms for de novo biosynthesis and salvage of purines, and thus, a concomitant alteration in uric acid metabolism. Moreover, gut microbiota can produce substrates that might cross the portal vein so the liver can generate de novo purinogenic amino acids, as well as uric acid. Therefore, the extrarenal excretion of uric acid needs to be considered as a factor in gout development. Nevertheless, further studies are needed to fully understand the role of gut microbiome in uric acid production and its extrarenal excretion, and to point out possible bacteria or bacterial enzymes that could be used as probiotic coadjutant treatment in gout patients.
Collapse
Affiliation(s)
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Calzada México-Xochimilco 289, Arenal de Guadalupe, 14389, Mexico City, Mexico.
| |
Collapse
|