1
|
Wu T, Li Y, Liu Y, Chu CQ. Preclinical RA: How to halt its progression. Best Pract Res Clin Rheumatol 2024:102030. [PMID: 39721896 DOI: 10.1016/j.berh.2024.102030] [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: 12/04/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder with a complex pathogenesis that evolves through various stages before clinical symptoms emerge. This review outlines the natural history of RA, starting from genetic predisposition and environmental triggers to preclinical autoimmunity and subsequent joint inflammation. Key genetic factors interact with environmental elements like smoking and infections, producing autoantibodies such as anti-citrullinated protein antibodies (ACPA) and rheumatoid factor, which precede clinical manifestations by several years. The preclinical phases offer critical opportunities for intervention aiming at halting disease progression. Preventive strategies including lifestyle modifications, dietary interventions, and targeted immune modulation may halt the progression to clinical RA in those at-risk individuals.
Collapse
Affiliation(s)
- Tong Wu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Lecheng Hospital, Sichuan University, Boao, Hainan, 571435, China.
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, 97239, USA; Rheumatology Section, VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
2
|
Brown HA, Morris AL, Pudlo NA, Hopkins AE, Martens EC, Golob JL, Koropatkin NM. Acarbose impairs gut Bacteroides growth by targeting intracellular glucosidases. mBio 2024; 15:e0150624. [PMID: 39565129 PMCID: PMC11633381 DOI: 10.1128/mbio.01506-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/08/2024] [Indexed: 11/21/2024] Open
Abstract
Acarbose is a type 2 diabetes medicine that prevents dietary starch breakdown into glucose by inhibiting host amylase and glucosidase enzymes. Numerous gut species in the Bacteroides genus enzymatically break down starch and change in relative abundance within the gut microbiome in acarbose-treated individuals. To mechanistically explain this observation, we used two model starch-degrading Bacteroides, Bacteroides ovatus (Bo), and Bacteroides thetaiotaomicron (Bt). Bt growth on starch polysaccharides is severely impaired by acarbose, whereas Bo growth is much less affected by the drug. The Bacteroides use a starch utilization system (Sus) to grow on starch. We hypothesized that Bo and Bt Sus enzymes are differentially inhibited by acarbose. Instead, we discovered that although acarbose primarily targets the Sus periplasmic GH97 enzymes in both organisms, the drug affects starch processing at multiple other points. Acarbose competes for transport through the TonB-dependent SusC proteins and binds to the Sus transcriptional regulators. Furthermore, Bo expresses a non-Sus GH97 (BoGH97D) when grown in starch with acarbose. The Bt homolog, BtGH97H, is not expressed in the same conditions, nor can overexpression of BoGH97D complement the Bt growth inhibition in the presence of acarbose. This work informs us about unexpected complexities of Sus function and regulation in Bacteroides, including variation between related species. Furthermore, this indicates that the gut microbiome may be a source of variable response to acarbose treatment for diabetes. IMPORTANCE Acarbose is a type 2 diabetes medication that works primarily by stopping starch breakdown into glucose in the small intestine. This is accomplished by the inhibition of host enzymes, leading to better blood sugar control via reduced ability to derive glucose from dietary starches. The drug and undigested starch travel to the large intestine where acarbose interferes with the ability of some bacteria to grow on starch. However, little is known about how gut bacteria interact with acarbose, including microbes that can use starch as a carbon source. Here, we show that two gut species, Bacteroides ovatus (Bo) and Bacteroides thetaiotaomicron (Bt), respond differently to acarbose: Bt growth is inhibited by acarbose, while Bo growth is less affected. We reveal a complex set of mechanisms involving differences in starch import and sensing behind the different Bo and Bt responses. This indicates the gut microbiome may be a source of variable response to acarbose treatment for diabetes via complex mechanisms in common gut microbes.
Collapse
Affiliation(s)
- Haley A. Brown
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Adeline L. Morris
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nicholas A. Pudlo
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ashley E. Hopkins
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Eric C. Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jonathan L. Golob
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nicole M. Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Cao Y, Chen J, Xiao J, Hong Y, Xu K, Zhu Y. Butyrate: a bridge between intestinal flora and rheumatoid arthritis. Front Immunol 2024; 15:1475529. [PMID: 39478858 PMCID: PMC11521846 DOI: 10.3389/fimmu.2024.1475529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
In patients with rheumatoid arthritis (RA), intestinal flora imbalance and butyrate metabolism disorders precede clinical arthritis and are associated with the pathogenesis of RA. This imbalance can alter the immunology and intestinal permeability of the intestinal mucosa, leading to damage to the intestinal barrier. In this context, bacteria and their metabolites can enter the bloodstream and reach the distant target tissues of the host, resulting in local inflammation and aggravating arthritis. Additionally, arthritis is also exacerbated by bone destruction and immune tolerance due to disturbed differentiation of osteoclasts and adaptive immune cells. Of note, butyrate is a metabolite of intestinal flora, which not only locally inhibits intestinal immunity and targets zonulin and tight junction proteins to alleviate intestinal barrier-mediated arthritis but also inhibits osteoclasts and autoantibodies and balances the immune responses of T and B lymphocytes throughout the body to repress bone erosion and inflammation. Therefore, butyrate is a key intermediate linking intestinal flora to the host. As a result, restoring the butyrate-producing capacity of intestinal flora and using exogenous butyrate are potential therapeutic strategies for RA in the future.
Collapse
Affiliation(s)
- Yang Cao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jingjing Chen
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jing Xiao
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yujie Hong
- Second Clinical College, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yan Zhu
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
4
|
Xu H, Luo Y, Li Q, Zhu H. Acupuncture influences multiple diseases by regulating gut microbiota. Front Cell Infect Microbiol 2024; 14:1371543. [PMID: 39040602 PMCID: PMC11260648 DOI: 10.3389/fcimb.2024.1371543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Acupuncture, an important green and side effect-free therapy in traditional Chinese medicine, is widely use both domestically and internationally. Acupuncture can interact with the gut microbiota and influence various diseases, including metabolic diseases, gastrointestinal diseases, mental disorders, nervous system diseases, and other diseases. This review presents a thorough analysis of these interactions and their impacts and examines the alterations in the gut microbiota and the potential clinical outcomes following acupuncture intervention to establish a basis for the future utilization of acupuncture in clinical treatments.
Collapse
Affiliation(s)
- Huimin Xu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaoqi Li
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Dalsgaard NB, Gasbjerg LS, Hansen LS, Nielsen DS, Rasmussen TS, Knop FK. Two weeks of acarbose treatment shows no effect on gut microbiome composition in patients with type 2 diabetes: a randomised, placebo-controlled, double-blind, crossover study. Endocr Connect 2024; 13:e240052. [PMID: 38842918 PMCID: PMC11227053 DOI: 10.1530/ec-24-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/06/2024] [Indexed: 06/07/2024]
Abstract
Aim The alpha-glucosidase inhibitor acarbose is approved for the treatment of type 2 diabetes (T2D). It acts in the lumen of the gut by reducing intestinal hydrolysis and absorption of ingested carbohydrates. This reduces postprandial blood glucose concentration and increases the content of carbohydrates in the distal parts of the intestine potentially influencing gut microbiome (GM) composition and possibly impacting the gut microbiome (GM) dysbiosis associated with T2D. Here, we investigated the effect of acarbose on GM composition in patients with T2D. Methods Faecal samples were collected in a previously conducted randomised, placebo-controlled, double-blind, crossover study in which 15 individuals with metformin-treated T2D (age 57-85 years, HbA1c 40-74 mmol/mol, BMI 23.6-34.6 kg/m2) were subjected to two 14-day treatment periods with acarbose and placebo, respectively, separated by a 6-week wash-out period. Faecal samples were collected before and by the end of each treatment period. The GM profiles were evaluated by 16S rRNA gene amplicon sequencing. Results The GM profiles after the treatment periods with acarbose or placebo remained unaffected (P > 0.7) when compared with the GM profiles before treatment. This applied to the analysis of within-sample diversity (α-diversity) and between-sample bacterial composition diversity (β-diversity). Additionally, no dominant bacterial species differentiated the treatment groups, and only minor increases in the relative abundances of Klebsiella spp. and Escherichia coli (P < 0.05) were observed after acarbose treatment. Conclusion In patients with metformin-treated T2D, 14 days of treatment with acarbose showed only minor effects on GM as seen in increased relative abundances of Klebsiella spp. and Escherichia coli.
Collapse
Affiliation(s)
- Niels B Dalsgaard
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Lærke S Gasbjerg
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Laura S Hansen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Dennis S Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Torben S Rasmussen
- Department of Food Science, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
6
|
Quesada S, Rosso AD, Mascardi F, Soler-Rivero V, Aguilera P, Mascuka SN, Boiro A, Arenielo E, Vijoditz G, Ferreyra-Mufarregue LR, Caputo MF, Cimolai MC, Coluccio Leskow F, Penas-Steinhardt A, Belforte FS. Integrative analysis of systemic lupus erythematosus biomarkers: Role of fecal hsa-mir-223-3p and gut microbiota in transkingdom dynamics. Mol Immunol 2024; 171:77-92. [PMID: 38795687 DOI: 10.1016/j.molimm.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/20/2024] [Accepted: 05/10/2024] [Indexed: 05/28/2024]
Abstract
Systemic lupus erythematosus (SLE) involves a florid set of clinical manifestations whose autoreactive origin is characterized by an overactivation of the immune system and the production of a large number of autoantibodies. Because it is a complex pathology with an inflammatory component, its pathogenesis is not yet fully understood, assuming both genetic and environmental predisposing factors. Currently, it is known that the role of the human microbiome is crucial in maintaining the transkingdom balance between commensal microorganisms and the immune system. In the present work we study the intestinal microbiota of Argentine patients with different stages of SLE receiving or not different treatments. Microbiota composition and fecal miRNAs were assessed by 16 S sequencing and qPCR. hsa-miR-223-3p, a miRNA involved in several inflammation regulation pathways, was found underexpressed in SLE patients without immunosuppressive treatment. In terms of microbiota there were clear differences in population structure (Weighted and Unweighted Unifrac distances, p-value <0.05) and core microbiome between cases and controls. In addition, Collinsella, Bifidobacterium, Streptococcus genera and aromatics degradation metabolisms were overrepresented in the SLE group. Medical treatment was also determinant as several microbial metabolic pathways were influenced by immunosuppressive therapy. Particularly, allantoin degradation metabolism was differentially expressed in the group of patients receiving immunosuppressants. Finally, we performed a logistic regression model (LASSO: least absolute shrinkage and selection operator) considering the expression levels of the fecal hsa-miR223-3p; the core microbiota; the differentially abundant bacterial taxa and the differentially abundant metabolic pathways (p<0.05). The model predicted that SLE patients could be associated with greater relative abundance of the formaldehyde oxidation pathway (RUMP_PWY). On the contrary, the preponderance of the ketodeoxyoctonate (Kdo) biosynthesis and activation route (PWY_1269) and the genera Lachnospiraceae_UCG_004, Lachnospira, Victivallis and UCG_003 (genus belonging to the family Oscillospiraceae of the class Clostridia) were associated with a control phenotype. Overall, the present work could contribute to the development of integral diagnostic tools for the comprehensive phenotyping of patients with SLE. In this sense, studying the commensal microbial profile and possible pathobionts associated with SLE in our population proposes more effective and precise strategies to explore possible treatments based on the microbiota of SLE patients.
Collapse
Affiliation(s)
- Sofía Quesada
- Laboratorio de Genómica Computacional (GeC-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Ayelén Daiana Rosso
- Laboratorio de Genómica Computacional (GeC-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina; Instituto de Ecología y Desarrollo Sustentable (INEDES-CONICET-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina
| | - Florencia Mascardi
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina; Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET, Instituto Universitario del Hospital Italiano (IUHI), Hospital Italiano de Buenos Aires (HIBA), Buenos Aires, Argentina
| | - Valeria Soler-Rivero
- Laboratorio de Genómica Computacional (GeC-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina
| | - Pablo Aguilera
- Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Sebastian Nicolas Mascuka
- Laboratorio de Genómica Computacional (GeC-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina
| | - Andrea Boiro
- Laboratorio de Genómica Computacional (GeC-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina
| | - Evangelina Arenielo
- Sección Inmunología, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Gustavo Vijoditz
- Sección Inmunología, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | | | - Marina Flavia Caputo
- Sección Inmunología, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - María Cecilia Cimolai
- Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina
| | - Federico Coluccio Leskow
- Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Alberto Penas-Steinhardt
- Laboratorio de Genómica Computacional (GeC-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina; Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Ciudad Autónoma de Buenos Aires, Argentina
| | - Fiorella Sabrina Belforte
- Laboratorio de Genómica Computacional (GeC-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Programa del Estudio de Comunicación y Señalización Interreino (PECSI-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina; Instituto de Ecología y Desarrollo Sustentable (INEDES-CONICET-UNLu), Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina.
| |
Collapse
|
7
|
Zhao L, Liu M, Zheng K, Xiao Q, Yuan L, Wu C, Bao J. Fufang Duzheng tablet attenuates adjuvant rheumatoid arthritis by inhibiting arthritis inflammation and gut microbiota disturbance in rats. Heliyon 2024; 10:e32705. [PMID: 39183834 PMCID: PMC11341321 DOI: 10.1016/j.heliyon.2024.e32705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 08/27/2024] Open
Abstract
Objective To explore the treatment effect and potential mechanism on gut microbiota, nutrition, and metabolism of Fufang Duzheng Tablet (DZGP) on rheumatoid arthritis (RA). Methods Collagen-induced arthritis rats' models were established and divided into three groups: model control group (FK), DZGP group (FZ, 0.45 g/kg/d), and methotrexate group (FM, 1.35 mg/kg), which were treated by gavage for 28 days. The physiopathologic changes of joints and body weight in each group were recorded; the morphology of synovial and ankle tissues was observed by hematoxylin-eosin staining, and the level of serum TNF-α and IL-1β was tested by ELISA. UPLC/MS-MS and network pharmacological analysis were used to identify the serum components, and 16S rDNA sequencing analysis was applied to the intestinal contents of rats. Results DZGP treatment significantly alleviated arthritis symptoms, pathological manifestations, toe thickness, and TNF-α and IL-1β levels in RA rats. We identified 105 metabolites and 18 components in the serum of DZGP-group rats. The main therapeutic targets of DZGP for anti-RA were TP53, epidermal growth factor receptor, and AKT1. Molecular docking showed that there was good binding efficiency between core components and main targets. 16S rDNA sequencing showed that DZGP treatment regulated the structure of the gut microbiota. Conclusion DZGP showed a good anti-inflammatory effect on RA and played an important role in improving the structure of the gut microbiota in RA rats.
Collapse
Affiliation(s)
- Liming Zhao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Hubei Minzu University, 445000, Enshi, China
| | - Meilin Liu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Kai Zheng
- Forest Seedlings and Wildlife Protection Management Station of Enshi Tujia and Miao Autonomous Prefecture, 445000, Enshi, China
| | - Qiang Xiao
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Hubei Minzu University, 445000, Enshi, China
| | - Lin Yuan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, 445000, Enshi, China
| | - Chuanfang Wu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Jinku Bao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China
| |
Collapse
|
8
|
Ruiz-Saavedra S, Salazar N, Suárez A, Diaz Y, González Del Rey C, González S, de Los Reyes-Gavilán CG. Human fecal alpha-glucosidase activity and its relationship with gut microbiota profiles and early stages of intestinal mucosa damage. Anaerobe 2024; 87:102853. [PMID: 38614290 DOI: 10.1016/j.anaerobe.2024.102853] [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: 12/21/2023] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024]
Abstract
OBJECTIVES We investigated potential relationships among initial lesions of the intestinal mucosa, fecal enzymatic activities and microbiota profiles. METHODS Fecal samples from 54 volunteers were collected after recruitment among individuals participating in a colorectal cancer (CRC) screening program in our region (Northern Spain) or attending for consultation due to clinical symptoms; intestinal mucosa samples were resected during colonoscopy. Enzymatic activities were determined in fecal supernatants by a semi-quantitative method. The fecal microbiota composition was determined by 16S rRNA gene-based sequencing. The results were compared between samples from clinical diagnosis groups (controls and polyps), according with the type of polyp (hyperplastic polyps or conventional adenomas) and considering the grade of dysplasia for conventional adenomas (low and high grade dysplasia). RESULTS High levels of α-glucosidase activity were more frequent among samples from individuals diagnosed with intestinal polyps, reaching statistical significance for conventional adenomas and for low grade dysplasia adenomas when compared to controls. Regarding the microbiota profiles, higher abundance of Christensenellaceae_R-7 group and Oscillospiraceae_UCG-002 were found in fecal samples displaying low α-glucosidase activity as compared with those with higher activity as well as in controls with respect to conventional adenomas. A relationship was evidenced among intestinal mucosal lesions, gut glucosidase activities and intestinal microbiota profiles. CONCLUSIONS Our findings suggest a relationship among altered fecal α-glucosidase levels, the presence of intestinal mucosal lesions, which can be precursors of CRC, and shifts in defined microbial groups of the fecal microbiota.
Collapse
Affiliation(s)
- Sergio Ruiz-Saavedra
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain; Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain; Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Adolfo Suárez
- Digestive Service, Central University Hospital of Asturias (HUCA), Oviedo, Spain; Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ylenia Diaz
- Digestive Service, Carmen and Severo Ochoa Hospital, Cangas del Narcea, Spain
| | - Carmen González Del Rey
- Department of Anatomical Pathology, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Sonia González
- Department of Functional Biology, University of Oviedo, Oviedo, Spain; Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Clara G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain; Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.
| |
Collapse
|
9
|
Brown HA, Morris AL, Pudlo NA, Hopkins AE, Martens EC, Golob JL, Koropatkin NM. Acarbose Impairs Gut Bacteroides Growth by Targeting Intracellular GH97 Enzymes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595031. [PMID: 38826241 PMCID: PMC11142093 DOI: 10.1101/2024.05.20.595031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Acarbose is a type-2 diabetes medicine that inhibits dietary starch breakdown into glucose by inhibiting host amylase and glucosidase enzymes. Numerous gut species in the Bacteroides genus enzymatically break down starch and change in relative abundance within the gut microbiome in acarbose-treated individuals. To mechanistically explain this observation, we used two model starch-degrading Bacteroides, Bacteroides ovatus (Bo) and Bacteroides thetaiotaomicron (Bt). Bt growth is severely impaired by acarbose whereas Bo growth is not. The Bacteroides use a starch utilization system (Sus) to grow on starch. We hypothesized that Bo and Bt Sus enzymes are differentially inhibited by acarbose. Instead, we discovered that although acarbose primarily targets the Sus periplasmic GH97 enzymes in both organisms, the drug affects starch processing at multiple other points. Acarbose competes for transport through the Sus beta-barrel proteins and binds to the Sus transcriptional regulators. Further, Bo expresses a non-Sus GH97 (BoGH97D) when grown in starch with acarbose. The Bt homolog, BtGH97H, is not expressed in the same conditions, nor can overexpression of BoGH97D complement the Bt growth inhibition in the presence of acarbose. This work informs us about unexpected complexities of Sus function and regulation in Bacteroides, including variation between related species. Further, this indicates that the gut microbiome may be a source of variable response to acarbose treatment for diabetes.
Collapse
Affiliation(s)
- Haley A. Brown
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Adeline L. Morris
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nicholas A. Pudlo
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ashley E. Hopkins
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Eric C. Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jonathan L. Golob
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nicole M. Koropatkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Chen T, Liu S, Zhang S, Song H, Zhuang Y, Ma J, Xiao J, Wang J, Ma Y, Wang Y, Wang W, Li S, Cao Z. Initial diet shapes resistance-gene composition and fecal microbiome dynamics in young ruminants during nursing. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 926:172103. [PMID: 38556024 DOI: 10.1016/j.scitotenv.2024.172103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
This study was conducted to examine how colostrum pasteurization affects resistance genes and microbial communities in calf feces. Forty female Holstein calves were randomly assigned to either the control (CON) group, which received unheated colostrum, or the pasteurized colostrum (PAT) group. The calves body weight was measured weekly before morning feeding. Calf starter intake were measured and recorded daily before morning feeding. Samples of colostrum were collected before feeding. Blood was collected on d 1 and 70 before morning feeding. Ten calves were randomly selected from each group (n = 20 calves total) for fecal sampling on d 3, 28, 56 and 70 for subsequent DNA extraction and metagenomic sequencing. Total bacterial counts in the colostrum were markedly higher in the CON group than in the PAT group. Pasteurized colostrum administration substantially reduced the ARO diversity and diminishes the abundance of Enterobacteriaceae, thereby decreasing their contribution to resistance genes. Pasteurization also reduced glucoside hydrolase-66 activity in 3-day-old calves which led to an increase in the activity of aminoglycoside antibiotics, resulting in 52.63 % of PAT-enriched bacteria acquiring aminoglycoside resistance genes. However, from the perspective of overall microbial community, the proportion of aminoglycoside, beta-lactam and tetracycline resistance genes carried by microbial community in PAT group was lower than CON group (P < 0.05). Fecal samples from the PAT group contained greater abundances of Subdoligranulum (P < 0.05) and Lachnospiraceae_NK4A136_group (P < 0.05) on days 28 and 70 compared to CON. Network analysis and abundance variations of the different bacteria obtained by linear discriminant analysis effect size analysis showed that pasteurized colostrum feeding reduced the interactions among related bacteria and maintained stability of the hind-gut microbiome. In conclusion, these findings underscore the intricate interactions between early diet, calf resistance-gene transmission and microbial dynamics, which should be carefully considered in calf-rearing practices.
Collapse
Affiliation(s)
- Tianyu Chen
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuai Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuyuan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Haotong Song
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yimin Zhuang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiaying Ma
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jianxin Xiao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jingjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yulin Ma
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yajing Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
11
|
Zhao H, Chen X, Sun Y, Shen P, Lin H, Sun F, Zhan S. Associations Between Thiazolidinediones Use and Incidence of Rheumatoid Arthritis: A Retrospective Population-Based Cohort Study. Arthritis Care Res (Hoboken) 2024; 76:486-496. [PMID: 38108108 DOI: 10.1002/acr.25277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/21/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVE Preclinical studies suggest that thiazolidinediones (TZDs) may have a protective effect on rheumatoid arthritis (RA), but evidence from population-based studies is scarce. This study aimed to assess the association between use of TZDs and incidence of RA in a retrospective cohort of patients with type 2 diabetes mellitus (T2DM). METHODS A retrospective cohort of patients with T2DM who were new users of TZDs or alpha glucosidase inhibitors (AGIs) was assembled. We applied the inverse probability of treatment weighted Cox model to estimate the hazard ratio (HR) of RA incidence associated with the use of TZDs compared with AGIs. RESULTS The final analysis included 56,796 new users of AGIs and 14,892 new users of TZDs. The incidence of RA was 187.4 and 135.2 per 100,000 person-years in AGI users and TZD users, respectively. Compared with use of AGIs, TZD use was associated with a reduction in RA incidence, with an HR of 0.72 (95% confidence interval [95% CI] 0.59-0.89). HRs for cumulative use of TZDs for 0.51 to 4.0 years and more than 4 years with incidence of RA were 0.55 (95% CI 0.35-0.88) and 0.74 (95% CI 0.57-0.98), respectively. Various subgroup analyses and sensitivity analyses were consistent with the primary analysis. CONCLUSION Use of TZDs is associated with a decreased risk of incident RA in patients with T2DM.
Collapse
Affiliation(s)
| | - Xiaowei Chen
- Peking University, Beijing, China, and Tianjin Medical University, Tianjin, China
| | - Yexiang Sun
- Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Peng Shen
- Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Hongbo Lin
- Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Feng Sun
- Peking University, Beijing, China
| | - Siyan Zhan
- Peking University and Peking University Third Hospital, Beijing, China
| |
Collapse
|
12
|
Song B, He J, Pan X, Kong L, Xiao C, Keerqin C, Song Z. Dietary Macleaya cordata extract supplementation improves the growth performance and gut health of broiler chickens with necrotic enteritis. J Anim Sci Biotechnol 2023; 14:113. [PMID: 37674220 PMCID: PMC10483844 DOI: 10.1186/s40104-023-00916-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/06/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND The poultry industry needs effective antibiotic alternatives to control outbreaks of necrotic enteritis (NE) caused by Clostridium perfringens. METHODS The aim of this study was to investigate the effects of dietary supplementation with Macleaya cordata extract (MCE) on the immune function and gut microbiota of broilers with NE. A total of 288 1-day-old broiler chicks were randomly assigned to a 2 × 2 factorial arrangement with two concentrations of dietary MCE supplementation (0 or 350 mg/kg of diet) and two disease challenge statuses (control or NE). RESULTS The results revealed that NE significantly increased the feed conversion rate (FCR), mortality, intestinal lesion score, the levels of IL-1β, IL-17 and IFN-γ/IL-4 in serum and IL-17/IL-10 in the jejunal mucosa, mRNA levels of TLR2, IFN-γ and pIgR in the jejunum, and Clostridium perfringens concentrations in the cecum. NE significantly decreased the body weight (BW), body weight gain (BWG), jejunal villus height, V/C, mRNA level of AMPK-α1 in jejunum, IL-4 level in the jejunal mucosa and lactic acid bacteria abundance in the cecum. MCE significantly increased BW, BWG, jejunal villus height, V/C, mRNA levels of occludin, ZO-1 and AMPK-α1 in the jejunum, the levels of IgA and IgG in serum and IL-10 in the jejunal mucosa and mRNA levels of NF-κB, IL-10 and MHC-II in the jejunum. Additionally, MCE significantly decreased the FCR, mortality, intestinal lesion score, jejunal crypt depth, the levels of IFN-γ and IL-17 in serum and IL-17/IL-10 in the jejunal mucosa, Clostridium perfringens concentrations in the cecum, and mRNA levels of IL-17/IL-10 in the jejunum. Moreover, NE significantly increased the abundance of bacteria that are associated with inflammation, obesity and depression (Alistipes, Barnesiella, Intestinimonas, RF39 and UCG-005) and significantly decreased the abundance of short-chain fatty acid (SCFA)-producing bacteria (Anaerotruncus, Butyricicoccus and Bacteroides) in the cecum. MCE significantly increased the abundance of SCFA-producing bacteria (Streptococcus, Ruminococcus_torques_group and Lachnospiraceae_NK4A136_group) and significantly reduced the abundance of bacteria that are associated with inflammation and obesity (Alistipes, Barnesiella and UCG-010) in the cecum. In the cecum of broilers with NE, the relative abundance of Barnesiella and Alistipes was higher and that of Lachnoclostridium and Shuttleworthia was lower. Interestingly, these trends were reversed by the addition of MCE to the diet. Spearman correlation analysis showed that Barnesiella and Alistipes were associated with enhanced intestinal inflammation and inhibited growth performance, whereas Lachnoclostridium and Shuttleworthia were associated with anti-inflammatory effects. CONCLUSIONS MCE ameliorated the loss of growth performance in broiler chickens with NE, probably by regulating the intestinal barrier, immune function, and gut microbiota.
Collapse
Affiliation(s)
- Bochen Song
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, Shandong, China
| | - Jie He
- Center for Mitochondria and Healthy Ageing, College of Life Sciences, Yantai University, Yantai, 264005, Shandong, China
| | - Xue Pan
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, Shandong, China
| | - Linglian Kong
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, Shandong, China
| | - Chuanpi Xiao
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, Shandong, China
- Precision Livestock and Nutrition Unit, University of Liège, Gembloux Agro-Bio TechGembloux, Belgium
| | - Chake Keerqin
- Phytobiotics (Jiangsu) Biotech Co., Ltd., Jintan, 213200, China
| | - Zhigang Song
- Key Laboratory of Efficient Utilization of Non-Grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, Shandong, China.
| |
Collapse
|
13
|
Koshida T, Gohda T, Sugimoto T, Asahara T, Asao R, Ohsawa I, Gotoh H, Murakoshi M, Suzuki Y, Yamashiro Y. Gut Microbiome and Microbiome-Derived Metabolites in Patients with End-Stage Kidney Disease. Int J Mol Sci 2023; 24:11456. [PMID: 37511232 PMCID: PMC10380578 DOI: 10.3390/ijms241411456] [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: 05/19/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The composition of the gut microbiome is altered in patients with chronic kidney disease (CKD). Dysbiosis leads to decreased levels of stool organic acids (OAs) and systemic inflammation, followed by accumulation of uremic toxins (UTs) and the development of end-stage kidney disease (ESKD). We assessed the relationship between the microbiome and UT levels or the development of ESKD by comparing patients undergoing hemodialysis (HD) and those with normal renal function (NRF). This cross-sectional study recruited 41 patients undergoing HD and 38 sex- and age-matched patients with NRF, and gut microbiome, levels of plasma UTs, inflammatory markers, and stool OAs were compared. The indices of beta-diversity differed significantly between patients with NRF and those undergoing HD, and between patients undergoing HD with and without type 2 diabetes. The levels of stool total OA, inflammatory markers, and UTs differed significantly between the patients with NRF and those undergoing HD. The combined main effects of type 2 diabetes and kidney function status were accumulation of indoxyl sulfate and p-cresyl sulfate. The relative abundances of Negativicutes and Megamonas were associated with development of ESKD and with the levels of UTs, even after adjustment for factors associated with the progression of ESKD. The present study indicates that the gut environment differs between patients with NRF and those undergoing HD and between patients undergoing HD with and without type 2 diabetes. Moreover, ESKD patients with diabetes accumulate more UTs derived from the gut microbiome, which might be associated with cardio-renal diseases and poor prognosis.
Collapse
Affiliation(s)
- Takeo Koshida
- Department of Nephrology, Faculty of Medicine, Juntendo University, Bunkyo-ku 113-8421, Tokyo, Japan
| | - Tomohito Gohda
- Department of Nephrology, Faculty of Medicine, Juntendo University, Bunkyo-ku 113-8421, Tokyo, Japan
| | - Takuya Sugimoto
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi-shi 186-0012, Tokyo, Japan
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi-shi 186-0012, Tokyo, Japan
| | - Rin Asao
- Department of Internal Medicine, Saiyu Soka Hospital, Soka-shi 340-0041, Saitama, Japan
| | - Isao Ohsawa
- Department of Internal Medicine, Saiyu Soka Hospital, Soka-shi 340-0041, Saitama, Japan
| | - Hiromichi Gotoh
- Department of Internal Medicine, Saiyu Soka Hospital, Soka-shi 340-0041, Saitama, Japan
| | - Maki Murakoshi
- Department of Nephrology, Faculty of Medicine, Juntendo University, Bunkyo-ku 113-8421, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Faculty of Medicine, Juntendo University, Bunkyo-ku 113-8421, Tokyo, Japan
| | - Yuichiro Yamashiro
- Probiotics Research Laboratory, Graduate School of Medicine, Juntendo University, Bunkyo-ku 113-8421, Tokyo, Japan
| |
Collapse
|
14
|
Tian J, Li C, Dong Z, Yang Y, Xing J, Yu P, Xin Y, Xu F, Wang L, Mu Y, Guo X, Sun Q, Zhao G, Gu Y, Qin G, Jiang W. Inactivation of the antidiabetic drug acarbose by human intestinal microbial-mediated degradation. Nat Metab 2023; 5:896-909. [PMID: 37157031 DOI: 10.1038/s42255-023-00796-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/30/2023] [Indexed: 05/10/2023]
Abstract
Drugs can be modified or degraded by the gut microbiota, which needs to be considered in personalized therapy. The clinical efficacy of the antidiabetic drug acarbose, an inhibitor of α-glucosidase, varies greatly among individuals for reasons that are largely unknown. Here we identify in the human gut acarbose-degrading bacteria, termed Klebsiella grimontii TD1, whose presence is associated with acarbose resistance in patients. Metagenomic analyses reveal that the abundance of K. grimontii TD1 is higher in patients with a weak response to acarbose and increases over time with acarbose treatment. In male diabetic mice, co-administration of K. grimontii TD1 reduces the hypoglycaemic effect of acarbose. Using induced transcriptome and protein profiling, we further identify an acarbose preferred glucosidase, Apg, in K. grimontii TD1, which can degrade acarbose into small molecules with loss of inhibitor function and is widely distributed in human intestinal microorganisms, especially in Klebsiella. Our results suggest that a comparatively large group of individuals could be at risk of acarbose resistance due to its degradation by intestinal bacteria, which may represent a clinically relevant example of non-antibiotic drug resistance.
Collapse
Affiliation(s)
- Jinzhong Tian
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, PR China
| | - Chong Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Zhixiang Dong
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, PR China
- University of Chinese Academy of Sciences, Beijing, PR China
| | - Yunpeng Yang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, PR China
| | - Jing Xing
- Lingang Laboratory, Shanghai, PR China
| | - Peijun Yu
- University of Chinese Academy of Sciences, Beijing, PR China
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, PR China
| | - Ying Xin
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Fengmei Xu
- Department of Endocrinology and Metabolism, Hebi Coal (group) Ltd. General Hospital, Hebi, PR China
| | - Lianwei Wang
- Department of Endocrinology and Metabolism, Zhumadian Central Hospital, Zhumadian, PR China
| | - Yahui Mu
- Department of Endocrinology and Metabolism, Huanghe Sanmenxia Hospital, Sanmenxia, PR China
| | - Xiangyang Guo
- Department of Endocrinology and Metabolism, Xinyang Central Hospital, Xinyang, PR China
| | - Qiang Sun
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, PR China
| | - Guoping Zhao
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, PR China
| | - Yang Gu
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, PR China.
| | - Guijun Qin
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
| | - Weihong Jiang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, PR China.
| |
Collapse
|
15
|
Woo AYM, Aguilar Ramos MA, Narayan R, Richards-Corke KC, Wang ML, Sandoval-Espinola WJ, Balskus EP. Targeting the human gut microbiome with small-molecule inhibitors. NATURE REVIEWS. CHEMISTRY 2023; 7:319-339. [PMID: 37117817 DOI: 10.1038/s41570-023-00471-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 04/30/2023]
Abstract
The human gut microbiome is a complex microbial community that is strongly linked to both host health and disease. However, the detailed molecular mechanisms underlying the effects of these microorganisms on host biology remain largely uncharacterized. The development of non-lethal, small-molecule inhibitors that target specific gut microbial activities enables a powerful but underutilized approach to studying the gut microbiome and a promising therapeutic strategy. In this Review, we will discuss the challenges of studying this microbial community, the historic use of small-molecule inhibitors in microbial ecology, and recent applications of this strategy. We also discuss the evidence suggesting that host-targeted drugs can affect the growth and metabolism of gut microbes. Finally, we address the issues of developing and implementing microbiome-targeted small-molecule inhibitors and define important future directions for this research.
Collapse
Affiliation(s)
- Amelia Y M Woo
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, MA, USA
| | | | - Rohan Narayan
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, MA, USA
| | | | - Michelle L Wang
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, MA, USA
| | - Walter J Sandoval-Espinola
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, MA, USA
- Universidad Nacional de Asunción, Facultad de Ciencias Exactas y Naturales, Departamento de Biotecnología, Laboratorio de Biotecnología Microbiana, San Lorenzo, Paraguay
| | - Emily P Balskus
- Harvard University, Department of Chemistry and Chemical Biology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
16
|
Śliżewska K, Włodarczyk M, Sobczak M, Barczyńska R, Kapuśniak J, Socha P, Wierzbicka-Rucińska A, Kotowska A. Comparison of the Activity of Fecal Enzymes and Concentration of SCFA in Healthy and Overweight Children. Nutrients 2023; 15:nu15040987. [PMID: 36839343 PMCID: PMC9966664 DOI: 10.3390/nu15040987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
In modern societies obesity has become a serious issue which must be urgently addressed. The health implications of neglected obesity are substantial, as not only does it affect individuals' everyday lives, but it also leads to significantly increased mortality due to the development of several disorders such as type-2 diabetes, cardiovascular diseases, cancers, and depression. The objective of this research was to investigate the alterations in selected health markers caused by overweight and obesity in children. The measured parameters were the activity of the fecal enzymes, the concentration of short-chain fatty acids (SCFAs), and the concentration of branched-chain fatty acids (BCFAs). The activity of the fecal enzymes, specifically α-glucosidase, α-galactosidase, β-glucosidase, β-galactosidase, and β-glucuronidase, was determined using spectrophotometry at a wavelength of 400 nm. Furthermore, concentrations of lactic acid, SCFAs (formic, acetic, propionic, butyric, and valeric acids), and BCFAs (isobutyric and isovaleric acids) were determined using the HPLC method. The obtained results reveal that obese children have different fecal enzyme activity and a different profile of fatty acids from children of normal weight. The group of obese children, when compared to children of normal weight, had increased concentrations of BCFAs (p < 0.05) and higher activity of potentially harmful enzymes such as β-glucosidase and β-glucuronidase (p < 0.05). In comparison, children of normal weight exhibited significantly increased concentrations of lactic acid and SCFAs (especially formic and butyric acids) (p < 0.05). Furthermore, their α-glucosidase and α-galactosidase activity were higher when compared to the group of obese children (p < 0.05). These results suggest that the prevalence of obesity has a significant impact on metabolites produced in the gastrointestinal tract, which might result in a higher chance of developing serious diseases.
Collapse
Affiliation(s)
- Katarzyna Śliżewska
- Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Technical University of Lodz, Wolczanska 171/173, 90-924 Łódź, Poland
- Correspondence: (K.Ś.); (M.W.)
| | - Michał Włodarczyk
- Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Technical University of Lodz, Wolczanska 171/173, 90-924 Łódź, Poland
- Correspondence: (K.Ś.); (M.W.)
| | - Martyna Sobczak
- Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Technical University of Lodz, Wolczanska 171/173, 90-924 Łódź, Poland
| | - Renata Barczyńska
- Department of Dietetics and Food Studies, Faculty of Science and Technology, Jan Dlugosz University, Armii Krajowej 13/15, 42-200 Czestochowa, Poland
| | - Janusz Kapuśniak
- Department of Dietetics and Food Studies, Faculty of Science and Technology, Jan Dlugosz University, Armii Krajowej 13/15, 42-200 Czestochowa, Poland
| | - Piotr Socha
- The Children’s Memorial Health Institute, aleja Dzieci Polskich 20, 04-736 Warsaw, Poland
| | | | - Aneta Kotowska
- The Children’s Memorial Health Institute, aleja Dzieci Polskich 20, 04-736 Warsaw, Poland
| |
Collapse
|
17
|
Romero-Figueroa MDS, Ramírez-Durán N, Montiel-Jarquín AJ, Horta-Baas G. Gut-joint axis: Gut dysbiosis can contribute to the onset of rheumatoid arthritis via multiple pathways. Front Cell Infect Microbiol 2023; 13:1092118. [PMID: 36779190 PMCID: PMC9911673 DOI: 10.3389/fcimb.2023.1092118] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/16/2023] [Indexed: 02/14/2023] Open
Abstract
Rheumatoid Arthritis (RA) is an autoimmune disease characterized by loss of immune tolerance and chronic inflammation. It is pathogenesis complex and includes interaction between genetic and environmental factors. Current evidence supports the hypothesis that gut dysbiosis may play the role of environmental triggers of arthritis in animals and humans. Progress in the understanding of the gut microbiome and RA. has been remarkable in the last decade. In vitro and in vivo experiments revealed that gut dysbiosis could shape the immune system and cause persistent immune inflammatory responses. Furthermore, gut dysbiosis could induce alterations in intestinal permeability, which have been found to predate arthritis onset. In contrast, metabolites derived from the intestinal microbiota have an immunomodulatory and anti-inflammatory effect. However, the precise underlying mechanisms by which gut dysbiosis induces the development of arthritis remain elusive. This review aimed to highlight the mechanisms by which gut dysbiosis could contribute to the pathogenesis of RA. The overall data showed that gut dysbiosis could contribute to RA pathogenesis by multiple pathways, including alterations in gut barrier function, molecular mimicry, gut dysbiosis influences the activation and the differentiation of innate and acquired immune cells, cross-talk between gut microbiota-derived metabolites and immune cells, and alterations in the microenvironment. The relative weight of each of these mechanisms in RA pathogenesis remains uncertain. Recent studies showed a substantial role for gut microbiota-derived metabolites pathway, especially butyrate, in the RA pathogenesis.
Collapse
Affiliation(s)
| | - Ninfa Ramírez-Durán
- Laboratory of Medical and Environmental Microbiology, Department of Medicine, Autonomous University of the State of Mexico, Toluca, Mexico
| | - Alvaro José Montiel-Jarquín
- Dirección de Educación e Investigación en Salud, Hospital de Especialidades de Puebla, Instituto Mexicano del Seguro Social, Puebla, Mexico
| | - Gabriel Horta-Baas
- Rheumatology Service, Internal Medicine Department, Instituto Mexicano del Seguro Social, Merida, Mexico
| |
Collapse
|
18
|
Chen S, Wang J, Dong N, Fang Q, Zhang Y, Chen C, Cui SW, Nie S. Polysaccharides from natural Cordyceps sinensis attenuated dextran sodium sulfate-induced colitis in C57BL/6J mice. Food Funct 2023; 14:720-733. [PMID: 36598450 DOI: 10.1039/d2fo02555h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
As potential candidates for treating ulcerative colitis (UC), polysaccharides have been attracting extensive interest in recent years. Cordyceps sinensis (C. sinensis) is a kind of traditional Chinese edible food, and its polysaccharide fractions have been found to be effective in regulating immunity and protecting the kidneys. To determine the potential function of polysaccharides from natural C. sinensis on UC, their effects in terms of histological, serological, biochemical, and immunological aspects on dextran sulphate sodium (DSS)-induced colitis mice model were investigated. Results showed that the polysaccharides significantly alleviated colitis by increasing the colon length, alleviating colon tissue damage, and inhibiting the activation of the NF-κB pathway. In addition, polysaccharides reduced the contents of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the serum, increased the number of goblet cells, and improved the expression of intestinal tight junction proteins (Occludin and Claudin-1). They also evidently enhanced the formation of IgA-secretory cells and sIgA contents. Furthermore, the polysaccharides modulated the gut microbiota by decreasing the relative abundance of Bilophila and increasing the relative abundance of Dehalobacterium, Coprococcus, Oscillospira, and Desulfovibrio, which is accompanied by an increase in the short chain fatty acids' (SCFAs) concentrations in cecal contents. These results suggested that C. sinensis polysaccharides possessed promising intervening effects on experimental acute UC in mice.
Collapse
Affiliation(s)
- Shuping Chen
- Jiangxi Key Laboratory of Natural Products and Functional Foods, Jiangxi Agricultural University, Nanchang 330045, China.,State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Junqiao Wang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Nan Dong
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Qiuyue Fang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Yanli Zhang
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Chunhua Chen
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Steve W Cui
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.,Guelph Research and Development Centre, Agriculture and Agri-Food Canada, 93 Stone Road West, Guelph, Ontario, N1G 5C9, Canada
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| |
Collapse
|
19
|
Abstract
Diabetes represents one of the most significant, and rapidly escalating, global healthcare crises we face today. Diabetes already affects one-tenth of the world's adults-more than 537 million people, numbers that have tripled since 2000 and are estimated to reach 643 million by 2030. Type 2 diabetes (T2D), the most prevalent form, is a complex disease with numerous contributing factors, including genetics, epigenetics, diet, lifestyle, medication use, and socioeconomic factors. In addition, the gut microbiome has emerged as a significant potential contributing factor in T2D development and progression. Gut microbes and their metabolites strongly influence host metabolism and immune function, and are now known to contribute to vitamin biosynthesis, gut hormone production, satiety, maintenance of gut barrier integrity, and protection against pathogens, as well as digestion and nutrient absorption. In turn, gut microbes are influenced by diet and lifestyle factors such as alcohol and medication use, including antibiotic use and the consumption of probiotics and prebiotics. Here we review current evidence regarding changes in microbial populations in T2D and the mechanisms by which gut microbes influence glucose metabolism and insulin resistance, including inflammation, gut permeability, and bile acid production. We also explore the interrelationships between gut microbes and different T2D medications and other interventions, including prebiotics, probiotics, and bariatric surgery. Lastly, we explore the particular role of the small bowel in digestion and metabolism and the importance of studying small bowel microbes directly in our search to find metabolically relevant biomarkers and therapeutic targets for T2D.
Collapse
Affiliation(s)
- Gillian M Barlow
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai, Los Angeles, CA, USA
| | - Ruchi Mathur
- Correspondence: Ruchi Mathur, MD, FRCPC, Director, Clinical Diabetes, Cedars-Sinai, 700 N San Vicente, Ste G271, West Hollywood, CA 90069, USA.
| |
Collapse
|
20
|
Shang J, Zhang Y, Guo R, Liu W, Zhang J, Yan G, Wu F, Cui W, Wang P, Zheng X, Wang T, Dong Y, Zhao J, Wang L, Xiao J, Zhao Z. Gut Microbiome Analysis Can Be Used as a Noninvasive Diagnostic Tool and Plays an Essential Role in the Onset of Membranous Nephropathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201581. [PMID: 35975460 PMCID: PMC9534961 DOI: 10.1002/advs.202201581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/06/2022] [Indexed: 05/15/2023]
Abstract
Membranous nephropathy (MN) is a common cause of nephrotic syndrome. The aim is to establish a non-invasive diagnostic model of MN using differential gut microbiome analysis, and to explore the relationship between the gut microbiome and MN pathogenesis in vivo. 825 fecal samples from MN patients and healthy participants are collected from multiple medical centers across China. Key operational taxonomic units (OTUs) obtained through 16S rRNA sequencing are used to establish a diagnostic model. A rat model of MN is developed to explore the relationship between the gut microbiome and the pathogenesis of MN. The diversity and richness of the gut microbiome are significantly lower in patients with MN than in healthy individuals. The diagnostic model based on seven OTUs achieves an excellent efficiency of 98.36% in the training group and also achieves high efficiency in cross-regional cohorts. In MN rat model, gut microbiome elimination prevents model establishment, but fecal microbiome transplantation restores the phenotype of protein urine. Gut microbiome analysis can be used as a non-invasive tool for MN diagnosis. The onset of MN depends on the presence of naturally colonized microbiome. Early intervention in the gut microbiome may help reduce urinary protein level in MN.
Collapse
Affiliation(s)
- Jin Shang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory Animal Platform of Academy of Medical SciencesZhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| | - Yiding Zhang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Ruixue Guo
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Wenli Liu
- Department of Clinical LaboratoryPeking Union Medical College HospitalPeking Union Medical College & China Academy of Medical ScienceBeijing100730China
| | - Jun Zhang
- Department of Nephrologythe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdong510630China
| | - Ge Yan
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Feng Wu
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Wen Cui
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Peipei Wang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Xuejun Zheng
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Ting Wang
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Yijun Dong
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Jing Zhao
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
| | - Li Wang
- Biobank of The First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| | - Jing Xiao
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| | - Zhanzheng Zhao
- Department of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
- Zhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory Animal Platform of Academy of Medical SciencesZhengzhou UniversityZhengzhouHenan450000P. R. China
- Laboratory of NephrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052P. R. China
| |
Collapse
|
21
|
Su YJ, Huang JY, Chu CQ, Wei JCC. Sulfonylureas or biguanides is associated with a lower risk of rheumatoid arthritis in patients with diabetes: A nationwide cohort study. Front Med (Lausanne) 2022; 9:934184. [PMID: 35966856 PMCID: PMC9363881 DOI: 10.3389/fmed.2022.934184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveDiabetes mellitus (DM) is associated with immune dysregulation, while sulfonylureas or biguanides have been linked to anti-inflammatory mechanisms. In this study, we aimed to examine the occurrence rate of rheumatoid arthritis (RA) among DM patients and its incidence rate between different treatments.MethodsThis cohort study used the Taiwan National Health Insurance Research Database between 1997 and 2013 to evaluate the primary outcomes of the preventive role of sulfonylureas or biguanides in the development of RA. We used the Chi-square test for categorical variables and Cox proportional hazard regression and log-rank test to explore the time for development of RA in DM patients. Logistic regression was adopted to estimate the odds ratio of RA in different dosages of medication exposure.ResultsOur cohort study included 94,141 DM cases. The risk of RA development of non-sulfonylureas/biguanides users among the DM group in each analysis was set as the reference, and the adjusted hazard ratio of RA in DM patients who were using sulfonylureas or biguanides was 0.73 (95% confidence interval 0.60–0.90). Within 1 year before the index date, compared with no-biguanides users, patients with more than 180 days of prescription of biguanides had a significantly lower RA risk. Similarly, the significantly lower risk of RA was still observed in DM patients who had more than 365 days of prescription of sulfonylurea within 2 or 3 years before the index date of first RA visit (all p < 0.05).ConclusionOur data suggest that sulfonylureas or biguanides are associated with a lower rate of RA development in patients with DM; the effect of biguanides appeared more rapid than that of sulfonylureas, but the sulfonylureas might have a longer effect on lowering RA development incidence.
Collapse
Affiliation(s)
- Yu-Jih Su
- Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jing-Yang Huang
- Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health and Science University and VA Portland Health Care System, Portland, OR, United States
- *Correspondence: Cong-Qiu Chu
| | - James Cheng-Chung Wei
- Department of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- James Cheng-Chung Wei
| |
Collapse
|
22
|
Zhou Y, Zhao H, Wang T, Zhao X, Wang J, Wang Q. Anti-Inflammatory and Anti-asthmatic Effects of TMDCT Decoction in Eosinophilic Asthma Through Treg/Th17 Balance. Front Pharmacol 2022; 13:819728. [PMID: 35211018 PMCID: PMC8861319 DOI: 10.3389/fphar.2022.819728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/24/2022] [Indexed: 12/28/2022] Open
Abstract
Tuo-Min-Ding-Chuan decoction (TMDCT) is a Traditional Chinese Medicine (TCM) formula consisting of twelve herbs that can relieve the symptoms and treat allergic asthma. Yet, the underlying mechanism of action is still unclear. In this study, we investigated the effect of TMDCT in regulating Treg/Th17 cells immune balance and explored potential metabolic and gut biomarkers associated with Treg and Th17 cells in eosinophilic asthma mice treated by TMDCT. We found that TMDCT increases Treg cells percentage and decreases Th17 cells percentage in the ovalbumin (OVA) -induced eosinophilic asthma mice model. Furthermore, Imidazoleacetic acid, dL-glutamine, L-pyroglutamic acid, 2-deoxy-d-glucose were preliminary identified as biomarkers in plasma metabolites treated by TMDCT, meanwhile genus Desulfovibrio, genus Butyricimonas and genus Prevotella 9 were preliminary identified as gut microbiota biomarkers after TMDCT treatment. These results provide an experimental foundation for the treatment of allergic asthma with Chinese herbal compounds.
Collapse
Affiliation(s)
- Yumei Zhou
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Haihong Zhao
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoshan Zhao
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ji Wang
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qi Wang
- National Institute of TCM Constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
23
|
Li N, Li X, Su R, Wu R, Niu HQ, Luo J, Gao C, Li X, Wang C. Low-Dose Interleukin-2 Altered Gut Microbiota and Ameliorated Collagen-Induced Arthritis. J Inflamm Res 2022; 15:1365-1379. [PMID: 35241924 PMCID: PMC8887675 DOI: 10.2147/jir.s344393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/26/2022] [Indexed: 12/20/2022] Open
Abstract
Purpose Low-dose interleukin-2 (ld-IL-2) has been shown to regulate the balance between effector T and regulatory T (Treg) cells and has been used in several clinical trials to treat autoimmune diseases including rheumatoid arthritis (RA). In this study, we investigated the effects of ld-IL-2 on collagen-induced arthritis (CIA) in mice. Methods Arthritis severity in CIA mice was measured using the arthritis index (AI), radiographs, and hematoxylin and eosin staining. Cytokines were detected using enzyme-linked immunosorbent assay. Gut microbiota alterations and short-chain fatty acid production were analyzed through 16S rRNA sequencing and gas chromatography. Results The AI scores of CIA mice treated with ld-IL-2 were significantly lower compared to the model group, which significantly reduced the severity of arthritis. Ld-IL-2 also altered the gut microbiota in CIA mice. The diversity, composition, and dominant species of gut microbiota were altered by ld-IL-2 treatment. Ld-IL-2 also increased short-chain fatty acid levels. There was a strong correlation between ld-IL-2 treatment and improved gut microbiota. Conclusion Ld-IL-2 significantly ameliorated joint inflammation and bone damage and improved gut microbial dysbiosis in CIA, indicating that it may be a promising therapy for RA patients.
Collapse
Affiliation(s)
- Na Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xuefei Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Hong-Qing Niu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Jing Luo
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women’s Hospital/Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Correspondence: Caihong Wang, Tel +8613603515399, Fax +863513365551, Email
| |
Collapse
|
24
|
Wang D, Liu J, Zhou L, Zhang Q, Li M, Xiao X. Effects of Oral Glucose-Lowering Agents on Gut Microbiota and Microbial Metabolites. Front Endocrinol (Lausanne) 2022; 13:905171. [PMID: 35909556 PMCID: PMC9326154 DOI: 10.3389/fendo.2022.905171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
The current research and existing facts indicate that type 2 diabetes mellitus (T2DM) is characterized by gut microbiota dysbiosis and disturbed microbial metabolites. Oral glucose-lowering drugs are reported with pleiotropic beneficial effects, including not only a decrease in glucose level but also weight loss, antihypertension, anti-inflammation, and cardiovascular protection, but the underlying mechanisms are still not clear. Evidence can be found showing that oral glucose-lowering drugs might modify the gut microbiome and thereby alter gastrointestinal metabolites to improve host health. Although the connections among gut microbial communities, microbial metabolites, and T2DM are complex, figuring out how antidiabetic agents shape the gut microbiome is vital for optimizing the treatment, meaningful for the instruction for probiotic therapy and gut microbiota transplantation in T2DM. In this review, we focused on the literatures in gut microbiota and its metabolite profile alterations beneficial from oral antidiabetic drugs, trying to provide implications for future study in the developing field of these drugs, such as combination therapies, pre- and probiotics intervention in T2DM, and subjects with pregestational diabetes and gestational diabetes mellitus.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Endocrinology, National Health Commission (NHC) Key Laboratory of Endocrinology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Jieying Liu
- Department of Endocrinology, National Health Commission (NHC) Key Laboratory of Endocrinology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
- Department of Medical Research Center, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Liyuan Zhou
- Department of Endocrinology, National Health Commission (NHC) Key Laboratory of Endocrinology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Qian Zhang
- Department of Endocrinology, National Health Commission (NHC) Key Laboratory of Endocrinology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Ming Li
- Department of Endocrinology, National Health Commission (NHC) Key Laboratory of Endocrinology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Xinhua Xiao
- Department of Endocrinology, National Health Commission (NHC) Key Laboratory of Endocrinology, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
- *Correspondence: Xinhua Xiao,
| |
Collapse
|
25
|
Zhang L, Chu CQ. Gut Microbiota-Medication Interaction in Rheumatic Diseases. Front Immunol 2021; 12:796865. [PMID: 34925383 PMCID: PMC8678121 DOI: 10.3389/fimmu.2021.796865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023] Open
Abstract
Besides its contribution to the development of rheumatic diseases, the gut microbiota interact with anti-rheumatic drugs. The intestinal microbiota can directly metabolize many drugs and indirectly change drug metabolism through a complex multi-dimensional interaction with the host, thus affecting individual response to drug therapy and adverse effects. The focus of the current review is to address recent advances and important progress in our understanding of how the gut microbiota interact with anti-rheumatic drugs and provide perspectives on promoting precision treatment, drug discovery, and better therapy for rheumatic diseases.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, United States.,Section of Rheumatology, Veterans Affairs (VA) Portland Healthcare System, Portland, OR, United States
| |
Collapse
|
26
|
Yang J, Xiong P, Bai L, Zhang Z, Zhou Y, Chen C, Xie Z, Xu Y, Chen M, Wang H, Zhu M, Yu J, Wang K. The Association of Altered Gut Microbiota and Intestinal Mucosal Barrier Integrity in Mice With Heroin Dependence. Front Nutr 2021; 8:765414. [PMID: 34805249 PMCID: PMC8600332 DOI: 10.3389/fnut.2021.765414] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
The gut microbiota is believed to play a significant role in psychological and gastrointestinal symptoms in heroin addicts. However, the underlying mechanism remains largely unknown. We show here that heroin addicts had a decrease in body mass index (BMI) and abnormal serum D-lactic acid (DLA), endotoxin (ET) and diamine oxidase (DAO) levels during their withdrawal stage, suggesting a potential intestinal injury. The gut microbial profiles in the mouse model with heroin dependence showed slightly decreased alpha diversity, as well as higher levels of Bifidobacterium and Sutterella and a decrease in Akkermansia at genus level compared to the control group. Fecal microbiota transplantation (FMT) further confirmed that the microbiota altered by heroin dependence was sufficient to impair body weight and intestinal mucosal barrier integrity in recipient mice. Moreover, short-chain fatty acids (SCFAs) profiling revealed that microbiota-derived propionic acid significantly decreased in heroin dependent mice compared to controls. Overall, our study shows that heroin dependence significantly altered gut microbiota and impaired intestinal mucosal barrier integrity in mice, highlighting the role of the gut microbiota in substance use disorders and the pathophysiology of withdrawal symptoms.
Collapse
Affiliation(s)
- Jiqing Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,Medical School, Kunming University of Science and Technology, Kunming, China.,National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pu Xiong
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ling Bai
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Zunyue Zhang
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong Zhou
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Cheng Chen
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhenrong Xie
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu Xu
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Gastrointestinal Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Minghui Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Huawei Wang
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mei Zhu
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Juehua Yu
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Centre for Experimental Studies and Research, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kunhua Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China.,National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Administrative Affairs, Yunnan University, Kunming, China
| |
Collapse
|
27
|
Bai H, Gu RJ, Chen LY, Qian Y, Yu ML, Xu SL, Xia XF, Liu YC, Zhang HR, Gu YH, Lu SF. Electroacupuncture interventions alleviates myocardial ischemia reperfusion injury through regulating gut microbiota in rats. Microvasc Res 2021; 138:104235. [PMID: 34453991 DOI: 10.1016/j.mvr.2021.104235] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/10/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022]
Abstract
Electroacupuncture (EA) intervention has a remarkable cardioprotection against myocardial ischemia reperfusion injury (MIRI). Recently, it has been suggested that the gut microbiota plays an important role in regulating the progression and prognosis of MIRI. The purpose of this study was to illustrate the relationship between gut microbiota and cardioprotection of EA on MIRI. We conducted a MIRI model by ligating the left anterior descending coronary artery for 30 min followed by reperfusion in male Sprague Dawley rats, which then received 7 days of EA intervention. Echocardiography was employed to evaluate left ventricular function. Fecal samples were collected for microbial analysis by 16S rDNA high-throughput sequencing. Blood samples and myocardium were collected for inflammatory cytokine detection by enzyme linked immunosorbent assay (ELISA) and Western blot. Hematoxylin & eosin (HE) staining and immunofluorescence of ileum tissue were performed for intestinal damage evaluation. After 7 days of EA intervention, the left ventricular function was improved with significantly increased ejection fraction and fractional shortening. Furthermore, we found that EA intervention reversed the changed gut microbiota induced by MIRI, including Clostridiales, RF39, S24-7, Desulfovibrio, and Allobaculum, improved the impaired gut barrier, reduced the production and circulation of lipopolysaccharide (LPS), inhibited the level of interleukin 6 (IL-6) and interleukin 12 (IL-12) in periphery and decreased the expression of Toll like receptor 4 (TLR4) and IL-6 in myocardium. EA intervention could improve the impaired gut mucosal barrier and reduce the production and circulation of LPS after MIRI through regulating gut microbiota, thus inhibiting the circulation and myocardium inflammation and finally exerted the cardioprotective effect.
Collapse
Affiliation(s)
- Hua Bai
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ren-Jun Gu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li-Yao Chen
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi Qian
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mei-Ling Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sen-Lei Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xue-Feng Xia
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu-Chen Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hong-Ru Zhang
- School of Basic Medical Sciences, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi-Huang Gu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sheng-Feng Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; Acupuncture and Tuina College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
28
|
Zhang L, Qing P, Yang H, Wu Y, Liu Y, Luo Y. Gut Microbiome and Metabolites in Systemic Lupus Erythematosus: Link, Mechanisms and Intervention. Front Immunol 2021; 12:686501. [PMID: 34335588 PMCID: PMC8319742 DOI: 10.3389/fimmu.2021.686501] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE), often considered the prototype of autoimmune diseases, is characterized by over-activation of the autoimmune system with abnormal functions of innate and adaptive immune cells and the production of a large number of autoantibodies against nuclear components. Given the highly complex and heterogeneous nature of SLE, the pathogenesis of this disease remains incompletely understood and is presumed to involve both genetic and environmental factors. Currently, disturbance of the gut microbiota has emerged as a novel player involved in the pathogenesis of SLE. With in-depth research, the understanding of the intestinal bacteria-host interaction in SLE is much more comprehensive. Recent years have also seen an increase in metabolomics studies in SLE with the attempt to identify potential biomarkers for diagnosis or disease activity monitoring. An intricate relationship between gut microbiome changes and metabolic alterations could help explain the mechanisms by which gut bacteria play roles in the pathogenesis of SLE. Here, we review the role of microbiota dysbiosis in the aetiology of SLE and how intestinal microbiota interact with the host metabolism axis. A proposed treatment strategy for SLE based on gut microbiome (GM) regulation is also discussed in this review. Increasing our understanding of gut microbiota and their function in lupus will provide us with novel opportunities to develop effective and precise diagnostic strategies and to explore potential microbiota-based treatments for patients with lupus.
Collapse
Affiliation(s)
- Lingshu Zhang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hang Yang
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yongkang Wu
- Department of Laboratory Medicine and Outpatient, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Horta-Baas G, Sandoval-Cabrera A, Romero-Figueroa MDS. Modification of Gut Microbiota in Inflammatory Arthritis: Highlights and Future Challenges. Curr Rheumatol Rep 2021; 23:67. [PMID: 34218340 DOI: 10.1007/s11926-021-01031-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW This Review evaluates the available information on the modification of the microbiota by diet, prebiotics, probiotics, or drugs and its association with the severity of arthritis in animals and humans and highlights how this modulation could have therapeutic applications in RA. RECENT FINDINGS The gut microbiota and microbiota-derived metabolites play a role in developing rheumatoid arthritis (RA) in animals and humans, making the intestinal microbiota an exciting novel approach to suppress autoimmunity. Studies in animal models of RA show that it is possible to modify the intestinal microbiota with drugs, natural products, diet, probiotics, and prebiotics. Furthermore, these changes showed beneficial effects on symptom relief in animal models of RA and that these effects were associated with modulation of the immune response. Therapies that modify the gut microbiota would significantly impact the preclinical stage of arthritis, based on the fact that dysbiosis occurs before clinical arthritis. The effects of interventions to modulate the microbiota could not reverse arthritis. Furthermore, the therapies modulating therapies in controlling symptoms were limited once arthritis developed. The results obtained in the study of acarbose, probiotics, and prebiotics suggest that these interventions may decrease the disease's incidence rather than treat or cure it.
Collapse
Affiliation(s)
- Gabriel Horta-Baas
- Servicio de Reumatología, Hospital General Regional número 1, Delegación Yucatán, Instituto Mexicano del Seguro Social, Calle 41 No. 439 x 34. Colonia Industrial, 97150, Mérida, Yucatán, Mexico.
| | - Antonio Sandoval-Cabrera
- Laboratorio de alta especialidad en Hemato-Oncología, Hospital para el Niño, IMIEM, Toluca, Mexico.,Facultad de Medicina, Campus Universitario Siglo XXl, Zinacantepec, State of Mexico, Mexico
| | - María Del Socorro Romero-Figueroa
- Facultad de Medicina, Campus Universitario Siglo XXl, Zinacantepec, State of Mexico, Mexico.,Centro de Investigación en Ciencias de la Salud, Campus Norte Huixquilucan, Universidad Anáhuac México, Mexico City, Mexico
| |
Collapse
|
30
|
Smith DL, Orlandella RM, Allison DB, Norian LA. Diabetes medications as potential calorie restriction mimetics-a focus on the alpha-glucosidase inhibitor acarbose. GeroScience 2021. [PMID: 33006707 DOI: 10.1007/s11357-020-00278-x/figures/1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023] Open
Abstract
The field of aging research has grown rapidly over the last half-century, with advancement of scientific technologies to interrogate mechanisms underlying the benefit of life-extending interventions like calorie restriction (CR). Coincident with this increase in knowledge has been the rise of obesity and type 2 diabetes (T2D), both associated with increased morbidity and mortality. Given the difficulty in practicing long-term CR, a search for compounds (CR mimetics) which could recapitulate the health and longevity benefits without requiring food intake reductions was proposed. Alpha-glucosidase inhibitors (AGIs) are compounds that function predominantly within the gastrointestinal tract to inhibit α-glucosidase and α-amylase enzymatic digestion of complex carbohydrates, delaying and decreasing monosaccharide uptake from the gut in the treatment of T2D. Acarbose, an AGI, has been shown in pre-clinical models to increase lifespan (greater longevity benefits in males), with decreased body weight gain independent of calorie intake reduction. The CR mimetic benefits of acarbose are further supported by clinical findings beyond T2D including the risk for other age-related diseases (e.g., cancer, cardiovascular). Open questions remain regarding the exclusivity of acarbose relative to other AGIs, potential off-target effects, and combination with other therapies for healthy aging and longevity extension. Given the promising results in pre-clinical models (even in the absence of T2D), a unique mechanism of action and multiple age-related reduced disease risks that have been reported with acarbose, support for clinical trials with acarbose focusing on aging-related outcomes and incorporating biological sex, age at treatment initiation, and T2D-dependence within the design is warranted.
Collapse
Affiliation(s)
- Daniel L Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1720 2nd Avenue S, Webb 423, Birmingham, AL, 35294-3360, USA.
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, USA.
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA.
- Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Rachael M Orlandella
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David B Allison
- School of Public Health, Indiana University - Bloomington, Bloomington, IN, USA
| | - Lyse A Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1720 2nd Avenue S, Webb 423, Birmingham, AL, 35294-3360, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
Smith DL, Orlandella RM, Allison DB, Norian LA. Diabetes medications as potential calorie restriction mimetics-a focus on the alpha-glucosidase inhibitor acarbose. GeroScience 2021; 43:1123-1133. [PMID: 33006707 PMCID: PMC8190416 DOI: 10.1007/s11357-020-00278-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
The field of aging research has grown rapidly over the last half-century, with advancement of scientific technologies to interrogate mechanisms underlying the benefit of life-extending interventions like calorie restriction (CR). Coincident with this increase in knowledge has been the rise of obesity and type 2 diabetes (T2D), both associated with increased morbidity and mortality. Given the difficulty in practicing long-term CR, a search for compounds (CR mimetics) which could recapitulate the health and longevity benefits without requiring food intake reductions was proposed. Alpha-glucosidase inhibitors (AGIs) are compounds that function predominantly within the gastrointestinal tract to inhibit α-glucosidase and α-amylase enzymatic digestion of complex carbohydrates, delaying and decreasing monosaccharide uptake from the gut in the treatment of T2D. Acarbose, an AGI, has been shown in pre-clinical models to increase lifespan (greater longevity benefits in males), with decreased body weight gain independent of calorie intake reduction. The CR mimetic benefits of acarbose are further supported by clinical findings beyond T2D including the risk for other age-related diseases (e.g., cancer, cardiovascular). Open questions remain regarding the exclusivity of acarbose relative to other AGIs, potential off-target effects, and combination with other therapies for healthy aging and longevity extension. Given the promising results in pre-clinical models (even in the absence of T2D), a unique mechanism of action and multiple age-related reduced disease risks that have been reported with acarbose, support for clinical trials with acarbose focusing on aging-related outcomes and incorporating biological sex, age at treatment initiation, and T2D-dependence within the design is warranted.
Collapse
Affiliation(s)
- Daniel L Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1720 2nd Avenue S, Webb 423, Birmingham, AL, 35294-3360, USA.
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, USA.
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA.
- Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Rachael M Orlandella
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David B Allison
- School of Public Health, Indiana University - Bloomington, Bloomington, IN, USA
| | - Lyse A Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, 1720 2nd Avenue S, Webb 423, Birmingham, AL, 35294-3360, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
32
|
Park JY, Kwon YW, Kim SA, Park SD, Kim CH, Kim JH, Lee JH. Polyherbal formula SC-E3 inhibits rheumatoid arthritis activity in a mouse model of type-II collagen-induced arthritis. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2020; 19:265-273. [PMID: 33349609 DOI: 10.1016/j.joim.2020.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE SC-E3 is a polyherbal formula that contains five medicinal herbs used frequently in traditional herbal medicine. In our previous study, we demonstrated the antioxidant and anti-inflammatory effects of SC-E3. The present study examined the effects of SC-E3 in a mouse model of type-II collagen-induced arthritis (CIA). METHODS In vivo, male DBA/1J mice were immunized by intradermal injection of bovine type-II collagen and complete or incomplete Freund's adjuvant, to induce arthritis. SC-E3 was orally administered daily for 23 days. In vitro, bone marrow-derived macrophages (BMMs) were treated with macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) in the absence or presence of SC-E3. RESULTS Administrations of SC-E3 were found to have anti-arthritic effects in the joints of CIA mice, as evidenced by reduced paw swelling, bone erosion and deformation, inflammatory cell infiltration, and inflammation in synovial membrane. SC-E3 also reduced serum levels of tumor necrosis factor-α, interleukin-1β, aspartate aminotransferase and alanine aminotransferase. Furthermore, tartrate-resistant acid phosphatase-positive osteoclast numbers in the joints were significantly lower in SC-E3-treated CIA mice than in CIA mice. In addition, the differentiations of BMMs to multinucleated osteoclasts induced by M-CSF and RANKL stimulation were dose-dependently reduced by SC-E3. CONCLUSION These results suggest that SC-E3 possesses substantial anti-arthritic activity because it inhibits pro-inflammatory cytokines and osteoclastogenesis, and that SC-E3 has potential therapeutic use for the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Ju-Yeon Park
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Young-Won Kwon
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Sun-Ah Kim
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Sun-Dong Park
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Chang-Hyun Kim
- Department of Medicine, College of Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Republic of Korea.
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea.
| |
Collapse
|