201
|
Xu Z, Tang H, Huang F, Qiao Z, Wang X, Yang C, Deng Q. Algal Oil Rich in n-3 PUFA Alleviates DSS-Induced Colitis via Regulation of Gut Microbiota and Restoration of Intestinal Barrier. Front Microbiol 2020; 11:615404. [PMID: 33391246 PMCID: PMC7772400 DOI: 10.3389/fmicb.2020.615404] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/26/2020] [Indexed: 12/18/2022] Open
Abstract
Algal oil is rich in docosahexaenoic acid (DHA) and has various health benefits against human metabolic disorders and disease. This study aimed to investigate the effects of DHA algal oil on colonic inflammation and intestinal microbiota in dextran sulfate sodium (DSS)-induced colitis mice model. Male C57BL/6 mice was induced colitis by 2.5% DSS and followed by 2 weeks of treatment with algal oil (250 or 500 mg/kg/day). The colonic inflammation was assessed by colon macroscopic damage scores, and the degree of neutrophil infiltration was evaluated by measuring tissue-associated myeloperoxidase (MPO) activity in colonic mucosa. Tight junction proteins in the colonic tissue were measured by real-time PCR and western blot. Moreover, the intestinal microbiota and shot chain fatty acids (SCFAs) were estimated by bioinformatic analysis and GC, respectively. Colonic damage due to DSS treatment was significantly ameliorated by algal oil supplementation. In addition, algal oil significantly inhibited the increases of malondialdehyde (MDA) content, MPO activity, pro-inflammatory cytokines level and tight junction proteins expression in DSS-treated mice. Furthermore, supplementation of algal oil modulated the intestinal microbiota structure in DSS induced colitis mice by increasing the proportion of the unidentified_S24_7 and decreasing the relative abundance of unidentified_Ruminococcaceae, Clostridium and Roseburia. On the analysis of SCFAs, the caecal content of acetic acid, propionic acid, isobutyric acid, buturic, and the total SCFAs showed a significant increase in algal oil-administered mice. Together, these results suggested that algal oil rich in DHA inhibited the progress of DSS-induced colitis in mice by modulating the intestinal microbiota and metabolites and repairing the intestinal barrier, which may be applied in the development of therapeutics for intestinal inflammation.
Collapse
Affiliation(s)
- Zhenxia Xu
- Oil Crops and Lipids Process Technology National and Local Joint Engineering Laboratory, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan, China
| | - Hu Tang
- Oil Crops and Lipids Process Technology National and Local Joint Engineering Laboratory, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan, China
| | - Fenghong Huang
- Oil Crops and Lipids Process Technology National and Local Joint Engineering Laboratory, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan, China
| | - Zhixian Qiao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xu Wang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Chen Yang
- Oil Crops and Lipids Process Technology National and Local Joint Engineering Laboratory, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan, China
| | - Qianchun Deng
- Oil Crops and Lipids Process Technology National and Local Joint Engineering Laboratory, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan, China
| |
Collapse
|
202
|
Suskind DL, Lee D, Kim YM, Wahbeh G, Singh N, Braly K, Nuding M, Nicora CD, Purvine SO, Lipton MS, Jansson JK, Nelson WC. The Specific Carbohydrate Diet and Diet Modification as Induction Therapy for Pediatric Crohn's Disease: A Randomized Diet Controlled Trial. Nutrients 2020; 12:nu12123749. [PMID: 33291229 PMCID: PMC7762109 DOI: 10.3390/nu12123749] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Crohn’s disease (CD) is a chronic inflammatory intestinal disorder associated with intestinal dysbiosis. Diet modulates the intestinal microbiome and therefore has a therapeutic potential. The aim of this study is to determine the potential efficacy of three versions of the specific carbohydrate diet (SCD) in active Crohn’s Disease. Methods: 18 patients with mild/moderate CD (PCDAI 15–45) aged 7 to 18 years were enrolled. Patients were randomized to either SCD, modified SCD(MSCD) or whole foods (WF) diet. Patients were evaluated at baseline, 2, 4, 8 and 12 weeks. PCDAI, inflammatory labs and multi-omics evaluations were assessed. Results: Mean age was 14.3 ± 2.9 years. At week 12, all participants (n = 10) who completed the study achieved clinical remission. The C-reactive protein decreased from 1.3 ± 0.7 at enrollment to 0.9 ± 0.5 at 12 weeks in the SCD group. In the MSCD group, the CRP decreased from 1.6 ± 1.1 at enrollment to 0.7 ± 0.1 at 12 weeks. In the WF group, the CRP decreased from 3.9 ± 4.3 at enrollment to 1.6 ± 1.3 at 12 weeks. In addition, the microbiome composition shifted in all patients across the study period. While the nature of the changes was largely patient specific, the predicted metabolic mode of the organisms increasing and decreasing in activity was consistent across patients. Conclusions: This study emphasizes the impact of diet in CD. Each diet had a positive effect on symptoms and inflammatory burden; the more exclusionary diets were associated with a better resolution of inflammation.
Collapse
Affiliation(s)
- David L. Suskind
- Department of Pediatrics, Division of Gastroenterology, Seattle Children’s Hospital and University of Washington, Seattle, WA 98105, USA; (D.L.); (G.W.); (N.S.); (K.B.); (M.N.)
- Correspondence: ; Tel.: +1-206-987-2521; Fax: +1-206-987-2721
| | - Dale Lee
- Department of Pediatrics, Division of Gastroenterology, Seattle Children’s Hospital and University of Washington, Seattle, WA 98105, USA; (D.L.); (G.W.); (N.S.); (K.B.); (M.N.)
| | - Young-Mo Kim
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Y.-M.K.); (C.D.N.); (J.K.J.); (W.C.N.)
| | - Ghassan Wahbeh
- Department of Pediatrics, Division of Gastroenterology, Seattle Children’s Hospital and University of Washington, Seattle, WA 98105, USA; (D.L.); (G.W.); (N.S.); (K.B.); (M.N.)
| | - Namita Singh
- Department of Pediatrics, Division of Gastroenterology, Seattle Children’s Hospital and University of Washington, Seattle, WA 98105, USA; (D.L.); (G.W.); (N.S.); (K.B.); (M.N.)
| | - Kimberly Braly
- Department of Pediatrics, Division of Gastroenterology, Seattle Children’s Hospital and University of Washington, Seattle, WA 98105, USA; (D.L.); (G.W.); (N.S.); (K.B.); (M.N.)
| | - Mason Nuding
- Department of Pediatrics, Division of Gastroenterology, Seattle Children’s Hospital and University of Washington, Seattle, WA 98105, USA; (D.L.); (G.W.); (N.S.); (K.B.); (M.N.)
| | - Carrie D. Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Y.-M.K.); (C.D.N.); (J.K.J.); (W.C.N.)
| | - Samuel O. Purvine
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (S.O.P.); (M.S.L.)
| | - Mary S. Lipton
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (S.O.P.); (M.S.L.)
| | - Janet K. Jansson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Y.-M.K.); (C.D.N.); (J.K.J.); (W.C.N.)
| | - William C. Nelson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (Y.-M.K.); (C.D.N.); (J.K.J.); (W.C.N.)
| |
Collapse
|
203
|
Selma-Royo M, Calatayud Arroyo M, García-Mantrana I, Parra-Llorca A, Escuriet R, Martínez-Costa C, Collado MC. Perinatal environment shapes microbiota colonization and infant growth: impact on host response and intestinal function. MICROBIOME 2020; 8:167. [PMID: 33228771 PMCID: PMC7685601 DOI: 10.1186/s40168-020-00940-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/14/2020] [Indexed: 05/11/2023]
Abstract
BACKGROUND Early microbial colonization triggers processes that result in intestinal maturation and immune priming. Perinatal factors, especially those associated with birth, including both mode and place of delivery are critical to shaping the infant gut microbiota with potential health consequences. METHODS Gut microbiota profile of 180 healthy infants (n = 23 born at home and n = 157 born in hospital, 41.7% via cesarean section [CS]) was analyzed by 16S rRNA gene sequencing at birth, 7 days, and 1 month of life. Breastfeeding habits and infant clinical data, including length, weight, and antibiotic exposure, were collected up to 18 months of life. Long-term personalized in vitro models of the intestinal epithelium and innate immune system were used to assess the link between gut microbiota composition, intestinal function, and immune response. RESULTS Microbiota profiles were shaped by the place and mode of delivery, and they had a distinct biological impact on the immune response and intestinal function in epithelial/immune cell models. Bacteroidetes and Bifidobacterium genus were decreased in C-section infants, who showed higher z-scores BMI and W/L during the first 18 months of life. Intestinal simulated epithelium had a stronger epithelial barrier function and intestinal maturation, alongside a higher immunological response (TLR4 route activation and pro-inflammatory cytokine release), when exposed to home-birth fecal supernatants, compared with CS. Distinct host response could be associated with different microbiota profiles. CONCLUSIONS Mode and place of birth influence the neonatal gut microbiota, likely shaping its interplay with the host through the maturation of the intestinal epithelium, regulation of the intestinal epithelial barrier, and control of the innate immune system during early life, which can affect the phenotypic responses linked to metabolic processes in infants. TRIAL REGISTRATION NCT03552939 . Video Abstract.
Collapse
Affiliation(s)
- M Selma-Royo
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain
| | - M Calatayud Arroyo
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain
| | - I García-Mantrana
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain
| | - A Parra-Llorca
- Neonatal Research Group, Health Research Institute La Fe, Valencia, Spain
| | - R Escuriet
- Gerencia de Procesos Integrales de Salud. Area Asistencial, Servicio Catalan de la Salud. Generalitat de Catalunya, Centre for Research in Health and Economics, Universidad Pompeu Fabra, Barcelona, Spain
| | - C Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, 46010, Valencia, Spain
- Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, INCLIVA, 46010, Valencia, Spain
| | - M C Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), Spanish National Research council, 46980, Valencia, Spain.
| |
Collapse
|
204
|
Yao Y, Feng Q, Shen J. Myosin light chain kinase regulates intestinal permeability of mucosal homeostasis in Crohn's disease. Expert Rev Clin Immunol 2020; 16:1127-1141. [PMID: 33183108 DOI: 10.1080/1744666x.2021.1850269] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Researchers have investigated the potential role of intestinal permeability in Crohn's disease pathogenesis. Intestinal permeability is usually mediated by cytoskeleton and intercellular junctions. The myosin light chain kinase (MLCK) is an enzyme that activates the myosin light chain to exert its function related to cytoskeleton contraction and tight junction regulation. The correlation between MLCK and Crohn's disease pathogenesis has been consistently proven. Areas covered: This study aims to expand the understanding of the regulation and function of MLCK in Crohn's disease. An extensive literature search in the MEDLINE database (via PubMed) has been performed up to Oct. 2020. The roles of MLCK in tight junction activation, intestinal permeability enhancement, and cell signal regulation are comprehensively discussed. Expert opinion: Targeting the MLCK-related pathways such as TNF-α in CD treatment has been put into clinical use. More accurate targeting such as MLCK and TNFR2 has been proposed to reduce side effects. MLCK may also have the potential to become biomarkers in fields like CD activity. With the application of cutting age research methods and tools, the MLCK research could be accelerated.
Collapse
Affiliation(s)
- Yiran Yao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center; Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University , Shanghai, China
| | - Qi Feng
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center; Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University , Shanghai, China
| |
Collapse
|
205
|
Caenepeel C, Sadat Seyed Tabib N, Vieira-Silva S, Vermeire S. Review article: how the intestinal microbiota may reflect disease activity and influence therapeutic outcome in inflammatory bowel disease. Aliment Pharmacol Ther 2020; 52:1453-1468. [PMID: 32969507 DOI: 10.1111/apt.16096] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/08/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal bacteria produce metabolites and by-products necessary for homeostasis. Imbalance in this equilibrium is linked to multiple pathologies including inflammatory bowel disease (IBD). The role of the gut microbiota in determining treatment response is becoming apparent, and may act as biomarker for efficacy. AIM To describe knowledge about the intestinal microbiota on disease severity and treatment outcomes in IBD METHODS: Descriptive review using PubMed to identify literature on the intestinal microbiota in IBD RESULTS: Severe IBD has a less diverse microbiota with fewer commensal microbiota communities and more opportunistic pathogenic bacteria originating from the oral cavity or respiratory tract. IBD treatments can alter gut microbiota composition, but in vitro/in vivo studies are needed to prove causation. A diversification of the microbiota is observed during remission. Patients with a more diverse baseline microbiome and higher microbial diversity show better response to anti-tumour necrosis factor-α, vedolizumab and ustekinumab therapy. Higher abundance of short chain fatty acid-producing bacteria, fewer mucus-colonising bacteria and lower abundance of pro-inflammatory bacteria have also been associated with a favourable outcome. Predictive models, based on a combination of microbiota, clinical data and serological markers, have good accuracy for treatment outcome and disease severity. CONCLUSION The intestinal microbiota in IBD carries a set of promising biomarkers of disease activity and prediction of therapeutic outcome. Current insights may also help in designing microbiota modulation strategies to improve outcomes in IBD.
Collapse
Affiliation(s)
| | | | - Sara Vieira-Silva
- Department of Microbiology and Immunology, Laboratory of Molecular Bacteriology, Rega Institute for Medical Research, VIB, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases & Metabolism, Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium.,Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
206
|
Simpson CA, Diaz-Arteche C, Eliby D, Schwartz OS, Simmons JG, Cowan CSM. The gut microbiota in anxiety and depression - A systematic review. Clin Psychol Rev 2020; 83:101943. [PMID: 33271426 DOI: 10.1016/j.cpr.2020.101943] [Citation(s) in RCA: 489] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/14/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022]
Abstract
Growing evidence indicates the community of microorganisms throughout the gastrointestinal tract, (i.e., gut microbiota), is associated with anxiety and depressive disorders. We present the first systematic review of the gut microbiota in anxiety disorders, along with an update in depression. Consideration of shared underlying features is essential due to the high rates of comorbidity. Systematic searches, following PRISMA guidelines, identified 26 studies (two case-control comparisons of the gut microbiota in generalised anxiety disorder, 18 in depression, one incorporating both anxiety/depression, and five including symptom-only measures). Alpha and beta diversity findings were inconsistent; however, differences in bacterial taxa indicated disorders may be characterised by a higher abundance of proinflammatory species (e.g., Enterobacteriaceae and Desulfovibrio), and lower short-chain fatty acid producing-bacteria (e.g., Faecalibacterium). Several taxa, and their mechanisms of action, may relate to anxiety and depression pathophysiology via communication of peripheral inflammation to the brain. Although the gut microbiota remains a promising target for prevention and therapy, future research should assess confounders, particularly diet and psychotropic medications, and should examine microorganism function.
Collapse
Affiliation(s)
- Carra A Simpson
- Melbourne School of Psychological Sciences, The University of Melbourne, VIC, Australia; Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, VIC, Australia.
| | - Carmela Diaz-Arteche
- Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, VIC, Australia
| | - Djamila Eliby
- Melbourne School of Psychological Sciences, The University of Melbourne, VIC, Australia; Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, VIC, Australia
| | - Orli S Schwartz
- Orygen, Centre for Youth Mental Health, The University of Melbourne, VIC, Australia
| | - Julian G Simmons
- Melbourne School of Psychological Sciences, The University of Melbourne, VIC, Australia; Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, VIC, Australia
| | | |
Collapse
|
207
|
Ota S, Sakuraba H, Hiraga H, Yoshida S, Satake M, Akemoto Y, Tanaka N, Watanabe R, Takato M, Murai Y, Ueno K, Niioka T, Hayakari M, Ishiguro Y, Fukuda S. Cyclosporine protects from intestinal epithelial injury by modulating butyrate uptake via upregulation of membrane monocarboxylate transporter 1 levels. Biochem Biophys Rep 2020; 24:100811. [PMID: 33102812 PMCID: PMC7578528 DOI: 10.1016/j.bbrep.2020.100811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 01/22/2023] Open
Abstract
Background and aims A relationship between treatment outcomes and intestinal microbiota in patients with inflammatory bowel diseases has been demonstrated. Cyclosporine treatment leads to rapid improvement in severe ulcerative colitis. We hypothesized that the potent effects of cyclosporine would be exerted through relationships between intestinal epithelial cells (IECs) and the host microbiota. The present study was designed to elucidate the effects of cyclosporine on monocarboxylate transporter 1 (MCT1) regulation and butyrate uptake by IECs. Methods Colitis was induced in C57BL6 mice via the administration of 4% dextran sulfate sodium in drinking water, following which body weights, colon lengths, and histological scores were evaluated. To examine the role of butyrate in the protective effects of cyclosporine, MCT1 inhibitor and an antibiotic cocktail was administered and tributyrin (TB; a prodrug of butyrate) was supplemented; MCT1 protein expression and acetylated histone 3 (AcH3) signals in IECs, as well as the MCT1-membrane fraction of Caco-2 cells, were evaluated. To explore butyrate uptake, as s butyrate derivatives, 3-bromopyruvic acid (3-BrPA) and 1-pyrenebutyric acid were used. Results Treatment with cyclosporine inhibited body weight loss and colon length shortening. However, treatment with MCT1 inhibitor and the antibiotic cocktail negated the efficacy of cyclosporine, whereas TB supplementation restored its protective effect. Furthermore, cyclosporine upregulated MCT1 expression in the membrane and the AcH3 signal in IECs, while also inducing higher anti-inflammatory cytokine production compared to that in the vehicle-treated mice. The transcription level of MCT1 mRNA in IECs and Caco-2 cells did not increase with cyclosporine treatment; however, cyclosporine treatment increased membrane MCT1 expression in these cells and uptake of butyrate derivative. Conclusion Cyclosporine treatment modulates butyrate uptake via the post-transcriptional upregulation of membrane MCT1 levels in IECs. The protective effect of cyclosporine needs microbiota-derived butyrate. Cyclosporine increased the fraction of MCT1 at the cell membrane. Cyclosporine enhanced butyrate uptake and regulatory cytokine expression.
Collapse
Affiliation(s)
- Shinji Ota
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hirotake Sakuraba
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hiroto Hiraga
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shukuko Yoshida
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.,Shibata Irika Co.Ltd.Hirosaki, Japan
| | - Miwa Satake
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yui Akemoto
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Nahoko Tanaka
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Rina Watanabe
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Maeda Takato
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasuhisa Murai
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kayo Ueno
- Division of Pharmaceutical Science, Hirosaki University Hospital, Hirosaki, Japan
| | - Takenori Niioka
- Division of Pharmaceutical Science, Hirosaki University Hospital, Hirosaki, Japan
| | - Makoto Hayakari
- Division of Pharmaceutical Science, Hirosaki University Hospital, Hirosaki, Japan
| | - Yoh Ishiguro
- Division of Clinical Research, Hirosaki National Hospital, National Hospital Organization, Hirosaki, Japan
| | - Shinsaku Fukuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
208
|
Schierová D, Březina J, Mrázek J, Fliegerová KO, Kvasnová S, Bajer L, Drastich P. Gut Microbiome Changes in Patients with Active Left-Sided Ulcerative Colitis after Fecal Microbiome Transplantation and Topical 5-aminosalicylic Acid Therapy. Cells 2020; 9:cells9102283. [PMID: 33066233 PMCID: PMC7602113 DOI: 10.3390/cells9102283] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease, and intestinal bacteria are implicated in the pathogenesis of this disorder. The administration of aminosalicylates (5-ASA) is a conventional treatment that targets the mucosa, while fecal microbial transplantation (FMT) is a novel treatment that directly targets the gut microbiota. The aim of this study was to identify changes in fecal bacterial composition after both types of treatments and evaluate clinical responses. Sixteen patients with active left-sided UC underwent enema treatment using 5-ASA (n = 8) or FMT (n = 8) with a stool from a single donor. Fecal microbiota were analyzed by 16S rDNA high-throughput sequencing, and clinical indices were used to assess the efficacy of treatments. 5-ASA therapy resulted in clinical remission in 50% (4/8) of patients, but no correlation with changes in fecal bacteria was observed. In FMT, remission was achieved in 37.5% (3/8) of patients and was associated with a significantly increased relative abundance of the families Lachnospiraceae, Ruminococcaceae, and Clostridiaceae of the phylum Firmicutes, and Bifidobacteriaceae and Coriobacteriaceae of the phylum Actinobacteria. At the genus level, Faecalibacterium, Blautia, Coriobacteria, Collinsela, Slackia, and Bifidobacterium were significantly more frequent in patients who reached clinical remission. However, the increased abundance of beneficial taxa was not a sufficient factor to achieve clinical improvement in all UC patients. Nevertheless, our preliminary results indicate that FMT as non-drug-using method is thought to be a promising treatment for UC patients.
Collapse
Affiliation(s)
- Dagmar Schierová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
- Correspondence: (D.S.); (J.M.); Tel.: +420-2-6709-0509 (D.S.); +420-2-6709-0506 (J.M.)
| | - Jan Březina
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| | - Jakub Mrázek
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
- Correspondence: (D.S.); (J.M.); Tel.: +420-2-6709-0509 (D.S.); +420-2-6709-0506 (J.M.)
| | - Kateřina Olša Fliegerová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
| | - Simona Kvasnová
- Institute of Animal Physiology and Genetics of the Czech Academy of Science, v.v.i., 142 20 Prague, Czech Republic; (K.O.F.); (S.K.)
| | - Lukáš Bajer
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| | - Pavel Drastich
- Hepatogastroenterology Department, Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (J.B.); (L.B.); (P.D.)
| |
Collapse
|
209
|
Zhu Y, Xiong Y, Gu Y, Li Q, Liu Y. Chiropractic Therapy Modulated Gut Microbiota and Attenuated Allergic Airway Inflammation in an Immature Rat Model. Med Sci Monit 2020; 26:e926039. [PMID: 32990279 PMCID: PMC7532697 DOI: 10.12659/msm.926039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background As a type of traditional Chinese massage, chiropractic therapy is applied to prevent and treat children with asthma in China. However, its mechanism of action is unclear. Allergic airway inflammation plays a key role in the occurrence and development of asthma, in which changes in gut microbiota are involved. The present study investigated the influence of chiropractic therapy on allergic airway inflammation (AAI) and gut microbiota in an immature rat model. Material/Methods Three-week-old male Sprague-Dawley rats were divided randomly into control (CN), AAI, and chiropractic (CP) groups. AAI and CP groups were sensitized and challenged with ovalbumin (OVA) to induce AAI. The CP group received chiropractic therapy during AAI modelling. AAI was assessed by cell counts in bronchoalveolar lavage fluid and HE staining of lung tissues. Plasma OVA-sIgE, IFN-γ, IL-4, and IL-10 levels were detected by ELISA. DNA extraction from feces samples was used for 16S rRNA gene sequencing and analyzed for gut microbiota by Quantitative Insights Into Microbial Ecology (QIIME). Results AAI group had significantly lower richness and diversity of gut microbiota along with Th2 response and allergic airway inflammation. Moreover, the AAI group had lower abundance of butyrate-producing bacterial taxa with more Lactobacillus. Chiropractic therapy significantly increased the richness and diversity of gut microbiota and increased butyrate-producing bacterial taxa and decreased Lactobacillus, along with attenuating Th2 response and allergic airway inflammation during AAI modelling. Conclusions Chiropractic therapy attenuated allergic airway inflammation and optimized gut microbiota in an immature rat model, which might promote the development of adult-like butyrogenic milieu, immunotolerance, and inflammation attenuation.
Collapse
Affiliation(s)
- Yan Zhu
- Graduate College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Ying Xiong
- Teaching and Research Section of Massage, Acupuncture and Massage College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Yun Gu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Acupuncture and Massage College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Qain Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Acupuncture and Massage College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Yu Liu
- Pediatric Massage Department, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
210
|
Assessing the Beneficial Effects of the Immunomodulatory Glycan LNFPIII on Gut Microbiota and Health in a Mouse Model of Gulf War Illness. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17197081. [PMID: 32992640 PMCID: PMC7579323 DOI: 10.3390/ijerph17197081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
The microbiota’s influence on host (patho) physiology has gained interest in the context of Gulf War Illness (GWI), a chronic disorder featuring dysregulation of the gut–brain–immune axis. This study examined short- and long-term effects of GWI-related chemicals on gut health and fecal microbiota and the potential benefits of Lacto-N-fucopentaose-III (LNFPIII) treatment in a GWI model. Male C57BL/6J mice were administered pyridostigmine bromide (PB; 0.7 mg/kg) and permethrin (PM; 200 mg/kg) for 10 days with concurrent LNFPIII treatment (35 μg/mouse) in a short-term study (12 days total) and delayed LNFPIII treatment (2×/week) beginning 4 months after 10 days of PB/PM exposure in a long-term study (9 months total). Fecal 16S rRNA sequencing was performed on all samples post-LNFPIII treatment to assess microbiota effects of GWI chemicals and acute/delayed LNFPIII administration. Although PB/PM did not affect species composition on a global scale, it affected specific taxa in both short- and long-term settings. PB/PM elicited more prominent long-term effects, notably, on the abundances of bacteria belonging to Lachnospiraceae and Ruminococcaceae families and the genus Allobaculum. LNFPIII improved a marker of gut health (i.e., decreased lipocalin-2) independent of GWI and, importantly, increased butyrate producers (e.g., Butyricoccus, Ruminococcous) in PB/PM-treated mice, indicating a positive selection pressure for these bacteria. Multiple operational taxonomic units correlated with aberrant behavior and lipocalin-2 in PB/PM samples; LNFPIII was modulatory. Overall, significant and lasting GWI effects occurred on specific microbiota and LNFPIII treatment was beneficial.
Collapse
|
211
|
In search for interplay between stool microRNAs, microbiota and short chain fatty acids in Crohn's disease - a preliminary study. BMC Gastroenterol 2020; 20:307. [PMID: 32958038 PMCID: PMC7507689 DOI: 10.1186/s12876-020-01444-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background Inflammatory bowel diseases are classic polygenic disorders, with genetic loads that reflect immunopathological processes in response to the intestinal microbiota. Herein we performed the multiomics analysis by combining the large scale surveys of gut bacterial community, stool microRNA (miRNA) and short chain fatty acid (SCFA) signatures to correlate their association with the activity of Crohn’s disease (CD). Methods DNA, miRNA, and metabolites were extracted from stool samples of 15 CD patients, eight with active disease and seven in remission, and nine healthy individuals. Microbial, miRNA and SCFA profiles were assessed using datasets from 16S rRNA sequencing, Nanostring miRNA and GC-MS targeted analysis, respectively. Results Pairwise comparisons showed that 9 and 23 taxa differed between controls and CD patients with active and inactive disease, respectively. Six taxa were common to both comparisons, whereas four taxa differed in CD patients. α-Diversity was lower in both CD groups than in controls. The levels of 13 miRNAs differed (p-value < 0.05; FC > 1.5) in CD patients and controls before FDR correction and 4 after. Of six SCFAs, the levels of two differed significantly (p-value < 0.05, FC > 1.5) in CD patients and controls, and the levels of four differed in patients with active and inactive CD. PLS-DA revealed models with smallest error rate for controls in bacterial component and inactive disease in metabolites. Conclusion A complex interrelationship may exist between gut dysbiosis, miRNA profiling and SCFA level in response to intestinal inflammation.
Collapse
|
212
|
Salamon D, Gosiewski T, Krawczyk A, Sroka-Oleksiak A, Duplaga M, Fyderek K, Kowalska-Duplaga K. Quantitative changes in selected bacteria in the stool during the treatment of Crohn's disease. Adv Med Sci 2020; 65:348-353. [PMID: 32590155 DOI: 10.1016/j.advms.2020.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/03/2020] [Accepted: 06/12/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE The aim of this study was to determine quantitative changes in selected species of bacteria (Bacteroides fragilis, Lactobacillus fermentum, Lactobacillus rhamnosus, Serratia marcescens) in the stool of patients with Crohn's disease (CD) in the course of induction treatment with exclusive enteral nutrition (EEN) or anti-tumor necrosis factor alpha (Infliximab, IFX) vs. healthy controls (HC). MATERIALS/METHODS DNA was isolated from stool samples of CD (n = 122) and HC (n = 17), and quantitative real-time Polymerase Chain Reaction (qPCR) was applied. In both treatment groups, the first stool sample was taken before the start of treatment, and the second 4 weeks after its end: in EEN (n = 48; age (mean; SD) 13.35 ± 3.09 years) and IFX groups (n = 13; age (mean; SD) 13.09 ± 3.76 years). RESULTS The only species that showed a statistically significant difference between the two groups of patients before any therapeutic intervention was L. fermentum. Moreover, its number increased after completion of EEN and differed significantly when compared with the HC. In the IFX group the number of L. fermentum decreased during the therapy but was significantly higher than in the HC. The number of S. marcescens in the EEN group was significantly lower than in the controls both before and after EEN. CONCLUSION The implemented treatment (EEN or IFX) modifies the microbiome in CD patients, but does not make it become the same as in HC.
Collapse
|
213
|
Ortigão R, Pimentel-Nunes P, Dinis-Ribeiro M, Libânio D. Gastrointestinal Microbiome - What We Need to Know in Clinical Practice. GE PORTUGUESE JOURNAL OF GASTROENTEROLOGY 2020; 27:336-351. [PMID: 32999906 PMCID: PMC7506249 DOI: 10.1159/000505036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/14/2019] [Indexed: 12/12/2022]
Abstract
Human gut microbiota plays an important role in individual health. When the balance between host and gut microbiota is disrupted, changes in microbiota composition and function occur, which is referred as dysbiosis. Environmental factors as diet, proton pump inhibitors, and antibiotics can lead to a permanent dysbiotic disruption. Clarification of these imbalances was made possible by recent advances in genome sequencing methods that supported acknowledgment of the interplay between microbiome and intestinal and extraintestinal disorders. This review focuses on the microbiota impact in inflammatory bowel disease, gastric cancer, colorectal cancer, nonalcoholic fatty liver disease (NAFLD), irritable bowel syndrome (IBS), and Clostridium difficile infection (CDI). Furthermore, novel therapies are summarized. Fecal microbiota transplant (FMT) is a successful and established therapy in recurrent CDI, and its application in other dysbiosis-related diseases is attracting enormous interest. Pre- and probiotics target microbial rebalance and have positive effects mainly in NAFLD, ulcerative colitis, IBS, and CDI patients. Promising anticarcinogenic effects have also been demonstrated in animal models. The literature increasingly describes microbial changes in many dysbiotic disorders and shows what needs to be treated. However, probiotics and FMT application in clinical practice suffers from a shortage of randomized controlled trials with standardized therapy regimens to support their recommendation.
Collapse
Affiliation(s)
- Raquel Ortigão
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal
| | - Pedro Pimentel-Nunes
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal
- MEDCIDS − Department of Community Medicine, Information and Decision in Health, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal
- MEDCIDS − Department of Community Medicine, Information and Decision in Health, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Diogo Libânio
- Department of Gastroenterology, Portuguese Oncology Institute of Porto, Porto, Portugal
- MEDCIDS − Department of Community Medicine, Information and Decision in Health, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
214
|
De Filippis F, Pasolli E, Ercolini D. The food-gut axis: lactic acid bacteria and their link to food, the gut microbiome and human health. FEMS Microbiol Rev 2020; 44:454-489. [PMID: 32556166 PMCID: PMC7391071 DOI: 10.1093/femsre/fuaa015] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Lactic acid bacteria (LAB) are present in foods, the environment and the animal gut, although fermented foods (FFs) are recognized as the primary niche of LAB activity. Several LAB strains have been studied for their health-promoting properties and are employed as probiotics. FFs are recognized for their potential beneficial effects, which we review in this article. They are also an important source of LAB, which are ingested daily upon FF consumption. In this review, we describe the diversity of LAB and their occurrence in food as well as the gut microbiome. We discuss the opportunities to study LAB diversity and functional properties by considering the availability of both genomic and metagenomic data in public repositories, as well as the different latest computational tools for data analysis. In addition, we discuss the role of LAB as potential probiotics by reporting the prevalence of key genomic features in public genomes and by surveying the outcomes of LAB use in clinical trials involving human subjects. Finally, we highlight the need for further studies aimed at improving our knowledge of the link between LAB-fermented foods and the human gut from the perspective of health promotion.
Collapse
Affiliation(s)
- Francesca De Filippis
- Department of Agricultural Sciences, University of Naples Federico II, via Università, 100, 80055, Portici (NA)Italy
- Task Force on Microbiome Studies, Corso Umberto I, 40, 80100, Napoli, Italy
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, via Università, 100, 80055, Portici (NA)Italy
- Task Force on Microbiome Studies, Corso Umberto I, 40, 80100, Napoli, Italy
| | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples Federico II, via Università, 100, 80055, Portici (NA)Italy
- Task Force on Microbiome Studies, Corso Umberto I, 40, 80100, Napoli, Italy
| |
Collapse
|
215
|
Sadeghi O, Milajerdi A, Siadat SD, Keshavarz SA, Sima AR, Vahedi H, Adibi P, Esmaillzadeh A. Effects of soy milk consumption on gut microbiota, inflammatory markers, and disease severity in patients with ulcerative colitis: a study protocol for a randomized clinical trial. Trials 2020; 21:565. [PMID: 32576228 PMCID: PMC7310397 DOI: 10.1186/s13063-020-04523-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Background Several strategies are recommended to alleviate clinical symptoms of ulcerative colitis (UC). Soy milk may affect UC through its anti-inflammatory properties. However, no study has examined the effects of soy milk consumption on gut microbiota and inflammatory biomarkers in patients with UC. The current study will be done to examine the effects of soy milk consumption on UC symptoms, inflammation, and gut microbiota in patients with UC. Methods This study is a randomized clinical trial, in which thirty patients with mild to moderate severity of UC will be randomly allocated to receive either 250 mL/day soy milk plus routine treatments (n = 15) or only routine treatments (n = 15) for 4 weeks. Assessment of anthropometric measures and biochemical indicators including serum concentrations of high-sensitivity C-reactive protein (hs-CRP), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interferon gamma (IFN-γ) will be done at the study baseline and end of trial. In addition, the quantity of butyrate-producing bacteria including Clostridium cluster IV, Faecalibacterium prausnitzii, and Roseburia spp.; prebiotic bacteria including Lactobacillus spp. and Bifidobacteria spp.; and mucus-degrading bacteria including Akkermansia muciniphila, Bacteroides fragilis, and Ruminococcus spp., as well as calprotectin and lactoferrin levels, will be explored in fecal samples. Also, the Firmicutes to Bacteroidetes ratio which is of significant relevance in human gut microbiota composition will be assessed. Discussion Altered gut microbiota has been reported as an important contributing factor to inflammation in patients with inflammatory bowel disease (IBD). Soy milk contains several components such as phytoestrogens with potential anti-inflammatory properties. This product might affect gut microbiota through its protein and fiber content. Therefore, soy milk might beneficially affect systemic inflammation, gut microbiota, and then clinical symptoms in patients with UC. Trial registration Iranian Registry of Clinical Trials (www.irct.ir) IRCT20181205041859N1. Registered on 27 January 2019.
Collapse
Affiliation(s)
- Omid Sadeghi
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box 14155-6117, Tehran, Iran
| | - Alireza Milajerdi
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box 14155-6117, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Ali Keshavarz
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Reza Sima
- Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Homayoon Vahedi
- Digestive Disease Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Adibi
- Integrative Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Esmaillzadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, P.O. Box 14155-6117, Tehran, Iran. .,Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
216
|
YAP Aggravates Inflammatory Bowel Disease by Regulating M1/M2 Macrophage Polarization and Gut Microbial Homeostasis. Cell Rep 2020; 27:1176-1189.e5. [PMID: 31018132 DOI: 10.1016/j.celrep.2019.03.028] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/17/2018] [Accepted: 03/07/2019] [Indexed: 12/30/2022] Open
Abstract
Inflammation, epithelial cell regeneration, macrophage polarization, and gut microbial homeostasis are critical for the pathological processes associated with inflammatory bowel disease (IBD). YAP (Yes-associated protein) is a key component of the Hippo pathway and was recently suggested to promote epithelial cell regeneration for IBD recovery. However, it is unclear how YAP regulates macrophage polarization, inflammation, and gut microbial homeostasis. Although YAP has been shown to promote epithelial regeneration and alleviate IBD, here we show that YAP in macrophages aggravates IBD, accompanied by the production of antimicrobial peptides and changes in gut microbiota. YAP impairs interleukin-4 (IL-4)/IL-13-induced M2 macrophage polarization while promoting lipopolysaccharide (LPS)/interferon γ (IFN-γ)-triggered M1 macrophage activation for IL-6 production. In addition, YAP expression is differently regulated during the induction of M2 versus M1 macrophages. This study suggests that fully understanding the multiple functions of YAP in different cell types is crucial for IBD therapy.
Collapse
|
217
|
Park SK, Kim HN, Choi CH, Im JP, Cha JM, Eun CS, Kim TO, Kang SB, Bang KB, Kim HG, Jung Y, Yoon H, Han DS, Lee CW, Ahn K, Kim HL, Park DI. Differentially Abundant Bacterial Taxa Associated with Prognostic Variables of Crohn's Disease: Results from the IMPACT Study. J Clin Med 2020; 9:E1748. [PMID: 32516912 PMCID: PMC7357029 DOI: 10.3390/jcm9061748] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/18/2022] Open
Abstract
Limited studies have examined the intestinal microbiota composition in relation to Crohn's disease (CD) prognosis. We analyzed the differences in microbial communities and relevant metabolic pathways associated with prognostic variables in patients with CD. We applied 16S rRNA gene sequencing to analyze a cohort of 1110 CD and healthy control (HC) fecal samples. We categorized patients with CD into good (CD-G), intermediate (CD-I) and poor (CD-P) prognosis groups, according to the history of using biologics and intestinal resection. Microbiota α-diversity decreased more in CD-P than CD-G and CD-I. Microbiota ß-diversity in CD-P differed from that in CD-G and CD-I. Thirteen genera and 10 species showed differential abundance between CD-G and CD-P groups. Escherichia coli (p = 0.001) and species Producta (p = 0.01) and genera Lactobacillus (p = 0.003) and Coprococcus (p = 0.01) consistently showed differences between CD-G and CD-P groups after adjusting for confounding variables. Functional profiling suggested that the microbial catabolic pathways and pathways related to enterobacterial common antigen and lipopolysaccharide biosynthesis were better represented in the CD-P group than in the CD-G group, and E. coli were the top contributors to these pathways. CD prognosis is associated with altered microbiota composition and decreased diversity, and E. coli might be causally involved in CD progression, and may have adapted to live in inflammatory environments.
Collapse
Affiliation(s)
- Soo-kyung Park
- Division of Gastroenterology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea;
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea; (H.-N.K.); (C.-W.L.)
- Inflammatory Bowel Disease Study Group of the Korean Association for the Study of Intestinal Diseases, Seoul 06193, Korea; (C.H.C.); (J.P.I.); (T.-O.K.); (H.Y.)
| | - Han-Na Kim
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea; (H.-N.K.); (C.-W.L.)
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul 03063, Korea
| | - Chang Hwan Choi
- Inflammatory Bowel Disease Study Group of the Korean Association for the Study of Intestinal Diseases, Seoul 06193, Korea; (C.H.C.); (J.P.I.); (T.-O.K.); (H.Y.)
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul 06973, Korea
| | - Jong Pil Im
- Inflammatory Bowel Disease Study Group of the Korean Association for the Study of Intestinal Diseases, Seoul 06193, Korea; (C.H.C.); (J.P.I.); (T.-O.K.); (H.Y.)
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jae Myung Cha
- Department of Internal Medicine, Kyung Hee University Hospital at Gang Dong, Kyung Hee University School of Medicine, Seoul 02447, Korea;
| | - Chang Soo Eun
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri 11923, Korea; (C.S.E.); (D.-S.H.)
| | - Tae-Oh Kim
- Inflammatory Bowel Disease Study Group of the Korean Association for the Study of Intestinal Diseases, Seoul 06193, Korea; (C.H.C.); (J.P.I.); (T.-O.K.); (H.Y.)
- Division of Gastroenterology, Department of Internal Medicine, Inje University Haeundae Paik Hospital, Busan 48108, Korea
| | - Sang-Bum Kang
- Division of Gastroenterology, Department of Internal medicine, Daejeon St. Mary’s Hospital, The Catholic University of Korea, Daejeon 34943, Korea;
| | - Ki Bae Bang
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan 31116, Korea;
| | - Hyun Gun Kim
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul Hospital, Seoul 04401, Korea;
| | - Yunho Jung
- Division of Gastroenterology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan Hospital, Cheonan 31151, Korea;
| | - Hyuk Yoon
- Inflammatory Bowel Disease Study Group of the Korean Association for the Study of Intestinal Diseases, Seoul 06193, Korea; (C.H.C.); (J.P.I.); (T.-O.K.); (H.Y.)
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Dong-Soo Han
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri 11923, Korea; (C.S.E.); (D.-S.H.)
| | - Chil-Woo Lee
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea; (H.-N.K.); (C.-W.L.)
| | - Kwangsung Ahn
- Functional Genome Institute, PDXen Biosystems Inc., Daejeon 34129, Korea;
| | - Hyung-Lae Kim
- Department of Biochemistry, School of Medicine, Ewha Womans University, Seoul 07804, Korea;
| | - Dong Il Park
- Division of Gastroenterology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea;
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea; (H.-N.K.); (C.-W.L.)
- Inflammatory Bowel Disease Study Group of the Korean Association for the Study of Intestinal Diseases, Seoul 06193, Korea; (C.H.C.); (J.P.I.); (T.-O.K.); (H.Y.)
| |
Collapse
|
218
|
Malinowski B, Wiciński M, Sokołowska MM, Hill NA, Szambelan M. The Rundown of Dietary Supplements and Their Effects on Inflammatory Bowel Disease-A Review. Nutrients 2020. [PMID: 32423084 DOI: 10.3390/nu12051423.pmid:32423084;pmcid:pmc7284960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Inflammatory bowel diseases, including Crohn's disease and ulcerative colitis, are a life-long, chronic, and relapsing problem affecting 11.2 million people worldwide. To date, there is pharmacological therapy to treat symptoms such as diarrhea, constipation, and abdominal cramping/pain. These medications also help to alleviate everyday discomfort; however, there are no curative therapies. Recent studies have investigated the combination of pharmacological treatment along with nutritional interventions to improve quality of life and risk of disease relapse. Dietary supplements, specifically probiotics, polyphenols, fibers, fatty acids and low fermentable oligosaccharide, disaccharide, monosaccharide, and polyol diets (FODMAP diets), have been closely looked at to determine their effect, if any, on the development of inflammatory bowel disease and its course of progression. Approximately 30 studies were carefully reviewed and analyzed to appreciate the value of these above-mentioned supplements and their influence on this gastrointestinal disease. After analysis, it has been demonstrated that by implementing fibers, polyphenols, and fatty acids, as well as keeping a low-saccharide diet for those patients with Crohn's disease and ulcerative colitis can improve quality of life and invoke clinical remission. Some polyphenols, specifically curcumin and resveratrol, have proved to decrease disease activity in studies reviewed. Although these studies have become a topic of recent interest, it would be of great value to doctors and patients alike, to continue in this direction of research and to improve the findings for best treatment substances and dosages. This would lead to increased quality of life and disease control leading to fewer complications in the future.
Collapse
Affiliation(s)
- Bartosz Malinowski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Maya M Sokołowska
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Nicholas A Hill
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| | - Monika Szambelan
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090 Bydgoszcz, Poland
| |
Collapse
|
219
|
Acute Effects of Butyrate on Induced Hyperpermeability and Tight Junction Protein Expression in Human Colonic Tissues. Biomolecules 2020; 10:biom10050766. [PMID: 32422994 PMCID: PMC7277647 DOI: 10.3390/biom10050766] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
Intact intestinal barrier function is essential for maintaining intestinal homeostasis. A dysfunctional intestinal barrier can lead to local and systemic inflammation through translocation of luminal antigens and has been associated with a range of health disorders. Butyrate, a short-chain fatty acid derived from microbial fermentation of dietary fibers in the colon, has been described as an intestinal barrier-strengthening agent, although mainly by using in vitro and animal models. This study aimed to investigate butyrate’s ability to prevent intestinal hyperpermeability, induced by the mast cell degranulator Compound 48/80 (C48/80), in human colonic tissues. Colonic biopsies were collected from 16 healthy subjects and intestinal permeability was assessed by Ussing chamber experiments. Furthermore, the expression levels of tight junction-related proteins were determined by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Pre-treatment with 5 mM butyrate or 25 mM butyrate did not protect the colonic tissue against induced paracellular or transcellular hyperpermeability, measured by FITC-dextran and horseradish peroxidase passage, respectively. Biopsies treated with 25 mM butyrate prior to stimulation with C48/80 showed a reduced expression of claudin 1. In conclusion, this translational ex vivo study did not demonstrate an acute protective effect of butyrate against a chemical insult to the intestinal barrier in healthy humans.
Collapse
|
220
|
Malinowski B, Wiciński M, Sokołowska MM, Hill NA, Szambelan M. The Rundown of Dietary Supplements and Their Effects on Inflammatory Bowel Disease-A Review. Nutrients 2020; 12:nu12051423. [PMID: 32423084 PMCID: PMC7284960 DOI: 10.3390/nu12051423] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel diseases, including Crohn's disease and ulcerative colitis, are a life-long, chronic, and relapsing problem affecting 11.2 million people worldwide. To date, there is pharmacological therapy to treat symptoms such as diarrhea, constipation, and abdominal cramping/pain. These medications also help to alleviate everyday discomfort; however, there are no curative therapies. Recent studies have investigated the combination of pharmacological treatment along with nutritional interventions to improve quality of life and risk of disease relapse. Dietary supplements, specifically probiotics, polyphenols, fibers, fatty acids and low fermentable oligosaccharide, disaccharide, monosaccharide, and polyol diets (FODMAP diets), have been closely looked at to determine their effect, if any, on the development of inflammatory bowel disease and its course of progression. Approximately 30 studies were carefully reviewed and analyzed to appreciate the value of these above-mentioned supplements and their influence on this gastrointestinal disease. After analysis, it has been demonstrated that by implementing fibers, polyphenols, and fatty acids, as well as keeping a low-saccharide diet for those patients with Crohn's disease and ulcerative colitis can improve quality of life and invoke clinical remission. Some polyphenols, specifically curcumin and resveratrol, have proved to decrease disease activity in studies reviewed. Although these studies have become a topic of recent interest, it would be of great value to doctors and patients alike, to continue in this direction of research and to improve the findings for best treatment substances and dosages. This would lead to increased quality of life and disease control leading to fewer complications in the future.
Collapse
|
221
|
Ye Z, Wu C, Zhang N, Du L, Cao Q, Huang X, Tang J, Wang Q, Li F, Zhou C, Xu Q, Xiong X, Kijlstra A, Qin N, Yang P. Altered gut microbiome composition in patients with Vogt-Koyanagi-Harada disease. Gut Microbes 2020; 11:539-555. [PMID: 31928124 PMCID: PMC7524263 DOI: 10.1080/19490976.2019.1700754] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Vogt-Koyanagi-Harada (VKH) disease is a multisystemic autoimmune disorder characterized by granulomatous panuveitis. Gut microbiome has been considered to play a role in the pathogenesis of this disease but whether the alternation of gut microbiome was involved is unclear. This study was set up to identify abnormalities of gut microbiome composition in VKH disease. RESULTS Depleted butyrate-producing bacteria, lactate-producing bacteria and methanogens as well as enriched Gram-negative bacteria were identified in the active VKH patients, as well as in VKH patients of Mix enterotype and Bacteroides enterotype. Changes of gut microbiome in the VKH patients were partially restored after an immunosuppressive treatment. The disease susceptibility genotype HLA-DRA was associated with Bacteroides sp.2.1.33B, Paraprevotella clara, Alistipes finegoldii and Eubacterium eligens. A microbial marker profile including 40 disease-associated species was established to differentiate patients from controls. Another microbial marker profile including 37 species was found to be associated with the response to treatment. An animal experiment showed that transfer of gut microbiome from VKH patients could significantly exacerbate disease activity clinically and pathologically in the recipient mice. CONCLUSION Our results revealed a distinct gut microbiome signature in VKH patients and showed an exacerbating effect of this gut microbiome on experimental autoimmune uveitis (EAU). We also developed two microbial marker profiles in differentiating VKH patients from healthy controls as well as predicting the effectiveness of treatment.
Collapse
Affiliation(s)
- Zi Ye
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Chunyan Wu
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China,Institute of Intestinal Diseases, Realbio Genomics Institute, Shanghai, China
| | - Ni Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Liping Du
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Qingfeng Cao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Xinyue Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Jihong Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Qingfeng Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Fuzhen Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunjiang Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Qian Xu
- Institute of Intestinal Diseases, Realbio Genomics Institute, Shanghai, China
| | - Xiao Xiong
- Institute of Intestinal Diseases, Realbio Genomics Institute, Shanghai, China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Nan Qin
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China,Institute of Intestinal Diseases, Realbio Genomics Institute, Shanghai, China,Nan Qin Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 301 Yanchang Middle Rd, Shanghai200072, China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing, China,CONTACT Peizeng Yang The First Affiliated Hospital of Chongqing Medical University, Youyi Road 1, Chongqing400016, P. R. China
| |
Collapse
|
222
|
Wang Y, Zou Y, Wang J, Ma H, Zhang B, Wang S. The Protective Effects of 2'-Fucosyllactose against E. Coli O157 Infection Are Mediated by the Regulation of Gut Microbiota and the Inhibition of Pathogen Adhesion. Nutrients 2020; 12:nu12051284. [PMID: 32369957 PMCID: PMC7282266 DOI: 10.3390/nu12051284] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022] Open
Abstract
As the richest component in human milk oligosaccharides (HMOs), 2’-fucosyllactose (2’-FL) can reduce the colonization of harmful microbiota in vivo, thus lowering the risk of infection; however, the mechanism for this is still unclear. In this study, a model of Escherichia coli O157 infection in healthy adult mice was established to explore the effect of 2’-FL intervention on E. coli O157 colonization and its protective effects on mice. The results showed that 2’-FL intake reduced E. coli O157 colonization in mice intestine by more than 90% (p < 0.001), and it also reduced intestinal inflammation, increased the content of fecal short-chain fatty acids, and enhanced intestinal barrier function. These beneficial effects were attributed to the increased expression of mucins such as MUC2 (increased by more than 20%, p < 0.001), and inhibition of E. coli O157 cell adhesion (about 30% reduction, p < 0.001), and were associated with the modulation of gut microbiota composition. 2’-FL significantly increased the abundance of Akkermansia, a potential probiotic, which may represent the fundamental means by which 2’-FL enhances the expression of mucin and reduces the colonization of harmful bacteria. The current study may support the use of 2’-FL in the prevention of foodborne pathogen infections in human.
Collapse
Affiliation(s)
- Yuanyifei Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of education, Tianjin University of Science and Technology, Tianjin 300457, China;
| | - Yan Zou
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.Z.); (J.W.); (H.M.); (B.Z.)
| | - Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.Z.); (J.W.); (H.M.); (B.Z.)
| | - Hui Ma
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.Z.); (J.W.); (H.M.); (B.Z.)
| | - Bowei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.Z.); (J.W.); (H.M.); (B.Z.)
| | - Shuo Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of education, Tianjin University of Science and Technology, Tianjin 300457, China;
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (Y.Z.); (J.W.); (H.M.); (B.Z.)
- Correspondence: ; Tel.: +86-22-85358445
| |
Collapse
|
223
|
Zhao J, Li G, Lu W, Huang S, Zhang Z. Dominant and Subordinate Relationship Formed by Repeated Social Encounters Alters Gut Microbiota in Greater Long-Tailed Hamsters. MICROBIAL ECOLOGY 2020; 79:998-1010. [PMID: 31807860 DOI: 10.1007/s00248-019-01462-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/08/2019] [Indexed: 06/10/2023]
Abstract
Social stress can dramatically influence the health of animals via communication between gut microbiota and the HPA system. However, this effect has been rarely investigated among different social ranked animals after chronic repeated social encounters. In this study, we evaluated changes and differences in microbiota among control, dominant, and subordinate male greater long-tailed hamsters (Tscherskia triton) over 28 successive days of repeated social encounter. Our results indicated that as compared with the control group, short-term repeated social encounters significantly altered fecal microbiota of subordinate hamsters, while chronic repeated social encounters altered colonic mucosa-associated microbiota of both dominant and subordinate hamsters. Fecal microbiota showed a transition in composition and diversity on day 2 for the subordinate group but on day 4 for the control and dominant groups under repeated encounters. Compared with their baseline, genus Lactobacillus increased in both dominant and subordinate groups, while genus Bifidobacterium increased in the subordinate group and genus Adlercreutzia increased in the dominant group. Our results suggest that chronic repeated social encounter can alter diversity and composition of gut microbiota of hamsters in both feces and colonic mucosa, but the latter performed better in reflecting the effects of chronic stress on microbiota in this species. Future studies should focus on elucidating how these microbiota alterations may affect animal behavior and fitness.
Collapse
Affiliation(s)
- Jidong Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Guoliang Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Wei Lu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Shuli Huang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
224
|
Silva NOE, Brito BBD, Silva FAFD, Santos MLC, Melo FFD. Probiotics in inflammatory bowel disease: Does it work? World J Meta-Anal 2020; 8:54-66. [DOI: 10.13105/wjma.v8.i2.54] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/26/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
|
225
|
Wu M, Wang F, Yang J, Li P, Yan D, Yang Y, Zhang W, Ren J, Zhang Z, Wang M. The responses of the gut microbiota to MBL deficiency. Mol Immunol 2020; 122:99-108. [PMID: 32330757 DOI: 10.1016/j.molimm.2020.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
Mannose-binding lectin (MBL) deficiency is a common innate immune system deficiency, and is associated with exacerbations and increased colonization of some pathogens. However, the response of the gut microbiota, a pivotal factor in host health, to MBL deficiency is not clear. In this study, MBL-/- and wild-type (WT) mice were generated by backcrossing from MBL-A and MBL-C knockout (KO) mice, and fecal samples were collected at different ages (4th, 8th, 12th, 19th and 27th weeks). The gut microbiota was analyzed by high-throughput sequencing with universal 16S rDNA primers (V3-V5 region). The results showed that structural segregation of the gut microbiota occurred at the 8th, 12th, 19th and 27th weeks of age, although there were no significant differences in alpha diversities between MBL-/- and WT mice at different ages. Impressively, in MBL-/- mice, Akkermansia (from the family Verrucomicrobiaceae) were decreased significantly, Lactobacillus (from the family Lactobacillaceae) abundances, Alistipes and Rikenella (both from the family of Rikenellaceae) were always enriched. Network analysis showed that more interactions existed in the gut microbiota from WT mice (33 nodes and 70 edges) than in the gut microbiota from MBL-/- mice (23 nodes and 40 edges). The 16S rDNA function prediction results indicated that the abundances of predicted genes in the "immune system disease", "metabolic disease" and "nucleotide metabolism" pathways were significantly increased in the MBL-/- mice. In conclusion, this study revealed that the gut microbiota changed in MBL deficient mice, especially at ages older than 4 weeks.
Collapse
Affiliation(s)
- Minna Wu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Fanping Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China
| | - Jingwen Yang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China
| | - Puze Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Dong Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yonghui Yang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China
| | - Wei Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Jie Ren
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Zhenchao Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Mingyong Wang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Immunoregulation and Molecular Diagnostics, Xinxiang 453003, China.
| |
Collapse
|
226
|
Soltys K, Stuchlikova M, Hlavaty T, Gaalova B, Budis J, Gazdarica J, Krajcovicova A, Zelinkova Z, Szemes T, Kuba D, Drahovska H, Turna J, Stuchlik S. Seasonal changes of circulating 25-hydroxyvitamin D correlate with the lower gut microbiome composition in inflammatory bowel disease patients. Sci Rep 2020; 10:6024. [PMID: 32265456 PMCID: PMC7138827 DOI: 10.1038/s41598-020-62811-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/18/2020] [Indexed: 12/24/2022] Open
Abstract
Higher probability of the development of Crohn's disease (CD) and ulcerative colitis (UC) as a possible consequence of the north-south gradient has been recently suggested. Living far north or south of the equator is manifested in fluctuation of vitamin D (vitD) levels depending on the season in both healthy and affected individuals. In the present study we investigate the possible link between the seasonal serum vitD level to the microbial composition of the lower gut of Inflammatory Bowel disease (IBD) patients using 16S rRNA sequencing. Decrease of serum vitD level in winter/spring season in a cohort of 35 UC patients and 39 CD patients was confirmed. Low gut microbiota composition of patients with IBD correlated with the serum level of 25(OH)D that directly coupled to seasonal variability of the sunshine in the central European countries. It is supposed to be related to increased abundance of Actinobacteria and Proteobacteria in UC and Actinobacteria, Fusobacteria, Firmicutes and Bacteroidetes in CD. In summer/autumn period, we observed a reduction in abundance of bacterial genera typical for inflammation like Eggerthella lenta, Fusobacterium spp., Bacteroides spp., Collinsella aerofaciens, Helicobacter spp., Rhodococcus spp., Faecalibacterium prausnitzii; and increased abundance of Pediococcus spp. and Clostridium spp. and of Escherichia/Shigella spp.
Collapse
Affiliation(s)
- Katarina Soltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia.
- Comenius University Science Park, Comenius University in Bratislava, Ilkovicova 8, 84104, Bratislava, Slovakia.
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia.
| | - Martina Stuchlikova
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia
- National Transplant Organization, Limbova 14, 83303, Bratislava, Slovakia
| | - Tibor Hlavaty
- Department of Internal Medicine, Faculty of Medicine, Division of Gastroenterology, Comenius University in Bratislava and University hospital Bratislava, Ruzinovska 6, 826 06, Bratislava, Slovakia
| | - Barbora Gaalova
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia
| | - Jaroslav Budis
- Department of Computer Science, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, Mlynska dolina F1, 842 48, Bratislava, Slovakia
| | - Juraj Gazdarica
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia
| | - Anna Krajcovicova
- Department of Internal Medicine, Faculty of Medicine, Division of Gastroenterology, Comenius University in Bratislava and University hospital Bratislava, Ruzinovska 6, 826 06, Bratislava, Slovakia
| | - Zuzana Zelinkova
- Department of Gastroenterology, St Michael Hospital, Bratislava, Slovakia
| | - Tomas Szemes
- Comenius University Science Park, Comenius University in Bratislava, Ilkovicova 8, 84104, Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia
| | - Daniel Kuba
- National Transplant Organization, Limbova 14, 83303, Bratislava, Slovakia
| | - Hana Drahovska
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia
| | - Jan Turna
- Comenius University Science Park, Comenius University in Bratislava, Ilkovicova 8, 84104, Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia
| | - Stanislav Stuchlik
- Comenius University Science Park, Comenius University in Bratislava, Ilkovicova 8, 84104, Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovicova 6, 84215, Bratislava, Slovakia
| |
Collapse
|
227
|
Abstract
While there are numerous medical comorbidities associated with ASD, gastrointestinal (GI) issues have a significant impact on quality of life for these individuals. Recent findings continue to support the relationship between the gut microbiome and both GI symptoms and behavior, but the heterogeneity within the autism spectrum requires in-depth clinical characterization of these clinical cohorts. Large, diverse, well-controlled studies in this area of research are still needed. Although there is still much to discover about the brain-gut-microbiome axis in ASD, microbially mediated therapies, specifically probiotics and fecal microbiota transplantation have shown promise in the treatment of GI symptoms in ASD, with potential benefit to the core behavioral symptoms of ASD as well. Future research and clinical trials must increasingly consider complex phenotypes in ASD in stratification of large datasets as well as in design of inclusion criteria for individual therapeutic interventions.
Collapse
Affiliation(s)
- Virginia Saurman
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Kara G. Margolis
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Ruth Ann Luna
- Department of Pathology and Immunology, Texas Children’s Microbiome Center, Baylor College of Medicine, Texas Children’s Hospital, Feigin Tower, 1102 Bates Avenue, Suite 955, Houston, TX 77030, USA
| |
Collapse
|
228
|
Pittayanon R, Lau JT, Leontiadis GI, Tse F, Yuan Y, Surette M, Moayyedi P. Differences in Gut Microbiota in Patients With vs Without Inflammatory Bowel Diseases: A Systematic Review. Gastroenterology 2020; 158:930-946.e1. [PMID: 31812509 DOI: 10.1053/j.gastro.2019.11.294] [Citation(s) in RCA: 378] [Impact Index Per Article: 75.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/05/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Altering the intestinal microbiota has been proposed as a treatment for inflammatory bowel diseases (IBDs), but there are no established associations between specific microbes and IBD. We performed a systematic review to identify frequent associations. METHODS We searched the MEDLINE, EMBASE, Cochrane Database of Systematic Reviews, and Cochrane Central Register of Controlled Trials databases, through April 2, 2018 for studies that compared intestinal microbiota (from fecal or colonic or ileal tissue samples) among patients (adult or pediatric) with IBD vs healthy individuals (controls). The primary outcome was difference in specific taxa in fecal or intestinal tissue samples from patients with IBD vs controls. We used the Newcastle-Ottawa scale to assess the quality of studies included in the review. RESULTS We identified 2631 citations; 48 studies from 45 articles were included in the analysis. Most studies evaluated adults with Crohn's disease or ulcerative colitis. All 3 studies of Christensenellaceae and Coriobacteriaceae and 6 of 11 studies of Faecalibacterium prausnitzii reported a decreased amount of those organisms compared with controls, whereas 2 studies each of Actinomyces, Veillonella, and Escherichia coli revealed an increased amount in patients with Crohn's disease. For patients with ulcerative colitis, Eubacterium rectale and Akkermansia were decreased in all 3 studies, whereas E coli was increased in 4 of 9 studies. The microbiota diversity was either decreased or not different in patients with IBD vs controls. Fewer than 50% of the studies stated comparable sexes and ages of cases and controls. CONCLUSIONS In a systematic review, we found evidence for differences in abundances of some bacteria in patients with IBD vs controls, but we cannot make conclusions due to inconsistent results and methods among studies. Further large-scale studies, with better methods of assessing microbe populations, are needed.
Collapse
Affiliation(s)
- Rapat Pittayanon
- Department of Medicine, Division of Gastroenterology and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada; Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; King Chulalongkorn Memorial Hospital, The Thai Red Cross, Bangkok, Thailand
| | - Jennifer T Lau
- Department of Medicine, Division of Gastroenterology and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Grigorios I Leontiadis
- Department of Medicine, Division of Gastroenterology and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Frances Tse
- Department of Medicine, Division of Gastroenterology and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Yuhong Yuan
- Department of Medicine, Division of Gastroenterology and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael Surette
- Department of Medicine, Division of Gastroenterology and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Paul Moayyedi
- Department of Medicine, Division of Gastroenterology and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
229
|
Hernandez-Sanabria E, Heiremans E, Calatayud Arroyo M, Props R, Leclercq L, Snoeys J, Van de Wiele T. Short-term supplementation of celecoxib-shifted butyrate production on a simulated model of the gut microbial ecosystem and ameliorated in vitro inflammation. NPJ Biofilms Microbiomes 2020; 6:9. [PMID: 32075981 PMCID: PMC7031363 DOI: 10.1038/s41522-020-0119-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/22/2020] [Indexed: 12/25/2022] Open
Abstract
Celecoxib has been effective in the prevention and treatment of chronic inflammatory disorders through inhibition of altered cyclooxygenase-2 (COX-2) pathways. Despite the benefits, continuous administration may increase risk of cardiovascular events. Understanding microbiome-drug-host interactions is fundamental for improving drug disposition and safety responses of colon-targeted formulations, but little information is available on the bidirectional interaction between individual microbiomes and celecoxib. Here, we conducted in vitro batch incubations of human faecal microbiota to obtain a mechanistic proof-of-concept of the short-term impact of celecoxib on activity and composition of colon bacterial communities. Celecoxib-exposed microbiota shifted metabolic activity and community composition, whereas total transcriptionally active bacterial population was not significantly changed. Butyrate production decreased by 50% in a donor-dependent manner, suggesting that celecoxib impacts in vitro fermentation. Microbiota-derived acetate has been associated with inhibition of cancer markers and our results suggest uptake of acetate for bacterial functions when celecoxib was supplied, which potentially favoured bacterial competition for acetyl-CoA. We further assessed whether colon microbiota modulates anti-inflammatory efficacy of celecoxib using a simplified inflammation model, and a novel in vitro simulation of the enterohepatic metabolism. Celecoxib was responsible for only 5% of the variance in bacterial community composition but celecoxib-exposed microbiota preserved barrier function and decreased concentrations of IL-8 and CXCL16 in a donor-dependent manner in our two models simulating gut inflammatory milieu. Our results suggest that celecoxib-microbiome-host interactions may not only elicit adaptations in community composition but also in microbiota functionality, and these may need to be considered for guaranteeing efficient COX-2 inhibition.
Collapse
Affiliation(s)
- Emma Hernandez-Sanabria
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium.
| | - Evelien Heiremans
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Marta Calatayud Arroyo
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Ruben Props
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Laurent Leclercq
- Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, Antwerp, 2340, Belgium
| | - Jan Snoeys
- Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, Beerse, Antwerp, 2340, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000, Ghent, Belgium.
| |
Collapse
|
230
|
Chen X, Zhang Z, Cui B, Jiang A, Tao H, Cheng S, Liu Y. Combination of Chronic Alcohol Consumption and High-Salt Intake Elicits Gut Microbial Alterations and Liver Steatosis in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1750-1759. [PMID: 31971384 DOI: 10.1021/acs.jafc.9b07368] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Alcohol is a globally well-established cause of fatty liver disease (FLD). Increased salt consumption is associated with an increased prevalence of adipocyte hypertrophy and liver injury. In this study, high dietary salt potentiated chronic alcohol-induced hepatic damage. We explored the physiological mechanism of alcoholic FLD in the gastrointestinal tract. Male C57BL/6J mice (8-week-old) were fed a high-salt diet (HSD; 4% NaCl) with or without chronic ethanol (CE) for 1 month. The fecal microbiota, serum biochemical indices, intestinal permeability, level of liver damage, and liver mitochondria were evaluated. The HSD, CE, and their combination (HSDE) significantly changed the gut microbiota's structure, and the HSDE mice contained more probiotic species (e.g., Bifidobacterium and Lactobacillus). The serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase levels were increased, and the lipid was accumulated in the liver tissues in the CE, HSD, and HSDE groups, which indicated liver damage, especially in the HSDE group. The increased intestinal permeability and mitochondrial dysfunction in the liver cells caused greater injury in the HSDE group than in the other groups. Thus, consuming HSD with alcohol contributes to FLD development and progression.
Collapse
Affiliation(s)
- Xiao Chen
- College of Food Science , South China Agricultural University , Guangzhou 510642 , China
| | - Zheng Zhang
- State Key Laboratory of Biobased Material and Green Papermaking , Qilu University of Technology, Shandong Academy of Sciences , Jinan 250000 , China
| | - Bo Cui
- State Key Laboratory of Biobased Material and Green Papermaking , Qilu University of Technology, Shandong Academy of Sciences , Jinan 250000 , China
| | - Aimin Jiang
- College of Food Science , South China Agricultural University , Guangzhou 510642 , China
| | - Haiteng Tao
- State Key Laboratory of Biobased Material and Green Papermaking , Qilu University of Technology, Shandong Academy of Sciences , Jinan 250000 , China
| | | | - Yong Liu
- Yucheng Maternal and Child Health Hospital , Dezhou 251200 , China
| |
Collapse
|
231
|
Composite probiotics alleviate type 2 diabetes by regulating intestinal microbiota and inducing GLP-1 secretion in db/db mice. Biomed Pharmacother 2020; 125:109914. [PMID: 32035395 DOI: 10.1016/j.biopha.2020.109914] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/10/2020] [Accepted: 01/12/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUD/AIM Previous studies have found that probiotic fermented camel milk has anti-diabetic effect by inducing (glucagon-like peptide-1) GLP-1 secretion. Probiotics are valuable in prevention and treatment of diabetes. As a result, our team islolated 14 probiotics from fermented camel milk. These probiotics have beneficial characteristics, but the possible anti-diabetic mechanisms remains unclear. The present study aimed to explore the possoble anti-diabetic mechanisms of 14 probiotics. METHODS C57BL/Ks mice were normal group. The db/db mice were randomized into five groups: model group, metformin group, liraglutide group, low-dose and high-dose probiotic group. Biochemical parameters were determined by the respective assay kits. The levels of the short-chain fatty acids (SCFAs) and microbiota were respectively determined by gas chromatography and qRT-PCR. HE staining and immunofluorescence were used for histomorphological observation. Quantitative PCR and western-blot were determined the gene and protein expression of Bax, Bcl-2, Caspase-3 and PI3K/AKT. RESULTS Probiotics significantly improved blood glucose and blood lipid parameters, as well as the morphological changes of pancreas, liver and kidney. Probiotics improved the gut barrier function through increasing the levels of SCFA-producing bacteria and SCFAs as well as the expression of claudin-1 and mucin-2, and decreasing Escherichia coli and LPS level. In additon, probiotics enhanced insulin secretion through glucose-triggered GLP-1 secretion by upregulating G protein-coupled receptor 43/41 (GPR43/41), proglucagon and proconvertase 1/3 activity. Forthermore, probiotics protected pancreas against apoptosis, which may be dependent on the upregulation of PI3K/AKT pathway. CONCLUSIONS The anti-diabetic effect of 14 probiotics in db/db mice seem to be related to an increase of SCFA-producing bacteria, the improvement of intestinal barrier function and the upregulation of GLP-1 production, and indicate these probiotics might be a good candidate to prevent and treat diabetes.
Collapse
|
232
|
Amorim C, Silvério SC, Cardoso BB, Alves JI, Pereira MA, Rodrigues LR. In vitro assessment of prebiotic properties of xylooligosaccharides produced by Bacillus subtilis 3610. Carbohydr Polym 2020; 229:115460. [DOI: 10.1016/j.carbpol.2019.115460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 10/25/2022]
|
233
|
Abstract
PURPOSE OF REVIEW Probiotics are promising remedial treatments for symptoms of small intestine (SI) diseases and promoters of overall good health. Probiotics play an important role in supporting a healthy SI microbiome (eubiosis), and in preventing establishment of unhealthy microbiota. SI eubiosis promotes optimal nutrient uptake, and optimal nutritional status maintains a healthy SI, reducing the likelihood of SI diseases. It is important to understand the advantages and limitations of probiotic therapies. RECENT FINDINGS Microbial dysbiosis decreases the capacity of the small bowel to utilize and absorb dietary compounds. In some studies, probiotic supplements containing lactic acid bacteria and Bifidobacterium have been demonstrated effective in supporting beneficial microbes in the SI while improving barrier integrity and reducing nutrient malabsorption and SI disease-related pathology. Strain-specific probiotic therapy may be a natural and effective approach to restoring SI barrier integrity and eubiosis, resulting in improved nutrient absorption and better health, including reducing the incidence of and severity of SI diseases.
Collapse
Affiliation(s)
- Taylor C Judkins
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr., Gainesville, FL, 32611, USA
| | - Douglas L Archer
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr., Gainesville, FL, 32611, USA
| | | | - Rebecca J Solch
- Food Science and Human Nutrition Department, University of Florida, 572 Newell Dr., Gainesville, FL, 32611, USA.
| |
Collapse
|
234
|
Yokoyama K, Tsuchiya N, Yamauchi R, Miyayama T, Uchida Y, Shibata K, Fukuda H, Umeda K, Takata K, Tanaka T, Inomata S, Morihara D, Takeyama Y, Shakado S, Sakisaka S, Hirai F. Exploratory Research on the Relationship between Human Gut Microbiota and Portal Hypertension. Intern Med 2020; 59:2089-2094. [PMID: 32879200 PMCID: PMC7516306 DOI: 10.2169/internalmedicine.4628-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objective The relationship between gut microbiota and portal hypertension remains unclear. We investigated the characteristics of the gut microbiota in portal hypertension patients with esophago-gastric varices and liver cirrhosis. Methods Thirty-six patients (12 patients with portal hypertension, 12 healthy controls, and 12 non-cirrhosis patients) were enrolled in this university hospital study. Intestinal bacteria and statistical analyses were performed up to the genus level using the terminal restriction fragment length polymorphism method targeting 16S ribosomal RNA genes, with diversified regions characterizing each bacterium. Results Levels of Lactobacillales were significantly higher (p=0.045) and those of Clostridium cluster IV significantly lower (p=0.014) in patients with portal hypertension than in other patients. This Clostridium cluster contains many butanoic acid-producing strains, including Ruminococcace and Faecalibacterium prausnitzii. Clostridium cluster IX levels were also significantly lower (p=0.045) in portal hypertension patients than in other patients. There are many strains of Clostridium that produce propionic acid, and the effects on the host and the function of these bacterial species in the human intestine remain unknown. Regarding the Bifidobacterium genus, which is supposed to decrease as a result of cirrhosis, no significant decrease was observed in this study. Conclusion In the present study, we provided information on the characteristics of the gut microbiota of portal hypertension patients with esophago-gastric varices due to liver cirrhosis. In the future, we aim to develop probiotic treatments following further analyses that include the species level, such as the intestinal flora analysis method and next-generation sequencers.
Collapse
Affiliation(s)
- Keiji Yokoyama
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Naoaki Tsuchiya
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Ryo Yamauchi
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Takashi Miyayama
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Yotaro Uchida
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Kumiko Shibata
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Hiromi Fukuda
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Kaoru Umeda
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Kazuhide Takata
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Takashi Tanaka
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Shinjiro Inomata
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Daisuke Morihara
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Yasuaki Takeyama
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Satoshi Shakado
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Shotaro Sakisaka
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| | - Fumihito Hirai
- Department of Gastroenterology and Medicine, Fukuoka University Faculty of Medicine, Japan
| |
Collapse
|
235
|
Pei LY, Ke YS, Zhao HH, Liu WZ, Wang L, Jia C, Shi MN, Fu QH, Cui J, Li SC. Regulatory effect of Garidisan on dysbiosis of the gut microbiota in the mouse model of ulcerative colitis induced by dextran sulfate sodium. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:329. [PMID: 31752807 PMCID: PMC6873523 DOI: 10.1186/s12906-019-2750-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Background Ulcerative colitis (UC) is a modern refractory disease, and its etiology has been difficult to discern. Studies have shown that UC is closely associated with the gut microbiota. Garidisan is composed of wild poppy and Artemisia frigida Willd and is commonly used for the treatment of UC in Inner Mongolia, China. In clinical settings, Garidisan has been found to treat UC effectively, with low recurrence. Previous studies have shown that Garidisan has a good therapeutic effect on mice with UC, but the therapeutic mechanism is still unclear. In this study, we investigated the regulatory effect of Garidisan on dysbiosis of the gut microbiota in a UC mouse model and explored the possible mechanism of the therapeutic effect of Garidisan on UC. Methods The UC mouse model was established by the dextran sulfate sodium (DSS) circulating free water drinking method, and the luminal contents were sampled under sterile conditions. High-throughput sequencing of the 16S rRNA gene V3 + V4 region of the luminal contents of the control group, model group, and Garidisan group was conducted, and clustering of operational taxonomic units (OTUs) and species annotation were performed. The differences in species composition and microbial community structure between individual groups of samples were analyzed using MetaStat, LefSe, rank sum test, and Bayesian causal network analysis. Results The UC mouse model was successfully established and the sequencing results were of adequate quality. There were significant differences in the diversity of luminal contents between the control group, model group, and Garidisan group, and the differences between groups were greater than those within any group. The therapeutic effect of Garidisan on UC is attributed to the direct effect on the Lachnospiraceae family of bacteria. Conclusion Garidisan has a good regulatory effect on the gut microbiota, and Lachnospiraceae could be an important direct target of Garidisan for the treatment of UC.
Collapse
|
236
|
Mima K, Sakamoto Y, Kosumi K, Ogata Y, Miyake K, Hiyoshi Y, Ishimoto T, Iwatsuki M, Baba Y, Iwagami S, Miyamoto Y, Yoshida N, Ogino S, Baba H. Mucosal cancer-associated microbes and anastomotic leakage after resection of colorectal carcinoma. Surg Oncol 2019; 32:63-68. [PMID: 31765952 DOI: 10.1016/j.suronc.2019.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 11/10/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Clinical and experimental evidence suggests that colorectal mucosal microbiota changes during colorectal carcinogenesis and may impair colorectal anastomotic wound healing. Thus, we hypothesized that amounts of colorectal cancer-associated microbes in colorectal tissue might be associated with anastomotic leakage after resection for colorectal carcinoma. METHODS We analyzed 256 fresh frozen tissues of colorectal cancer from patients who underwent elective colorectal resection and anastomosis. Amounts of colorectal cancer-associated microbes, including Fusobacterium nucleatum, Escherichia coli possessing the polyketide synthase (pks) gene cluster, Enterococcus faecalis, and Bifidobacterium genus, in colorectal cancer tissues were measured by quantitative polymerase chain reaction assay; we equally dichotomized positive cases (high versus low). Multivariable logistic regression analysis was conducted to assess associations of these microbes with anastomotic leakage, adjusting for patient and tumor characteristics, and surgery-related factors. RESULTS Fusobacterium nucleatum, pks-positive Escherichia coli, Enterococcus faecalis, and Bifidobacterium genus were detected in colorectal carcinoma tissue in 140 (54%), 94 (36%), 193 (75%), and 89 (35%) of 256 cases, respectively. Compared with Bifidobacterium genus-negative cases, Bifidobacterium genus-high cases were associated with an increased risk of anastomotic leakage (multivariable odds ratio, 3.96; 95% confidence interval, 1.50 to 10.51; Ptrend = 0.004). The association of Fusobacterium nucleatum, pks-positive Escherichia coli, or Enterococcus faecalis with anastomotic leakage was not statistically significant. CONCLUSIONS The amount of Bifidobacterium genus in colorectal tissue is associated with an increased risk of anastomotic leakage after resection for colorectal cancer. These findings need to be validated to target gastrointestinal microflora for the prevention of anastomotic leakage after colorectal resection.
Collapse
Affiliation(s)
- Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan; Department of Surgery, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Yuki Sakamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Keisuke Kosumi
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Yoko Ogata
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Keisuke Miyake
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yukiharu Hiyoshi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Shiro Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
237
|
Pellicano C, Leodori G, Innocenti GP, Gigante A, Rosato E. Microbiome, Autoimmune Diseases and HIV Infection: Friends or Foes? Nutrients 2019; 11:E2629. [PMID: 31684052 PMCID: PMC6893726 DOI: 10.3390/nu11112629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/25/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023] Open
Abstract
Several studies highlighted the importance of the interaction between microbiota and the immune system in the development and maintenance of the homeostasis of the human organism. Dysbiosis is associated with proinflammatory and pathological state-like metabolic diseases, autoimmune diseases and HIV infection. In this review, we discuss the current understanding of the possible role of dysbiosis in triggering and/or exacerbating symptoms of autoimmune diseases and HIV infection. There are no data about the influence of the microbiome on the development of autoimmune diseases during HIV infection. We can hypothesize that untreated patients may be more susceptible to the development of autoimmune diseases, due to the presence of dysbiosis. Eubiosis, re-established by probiotic administration, can be used to reduce triggers for autoimmune diseases in untreated HIV patients, although clinical studies are needed to evaluate the role of the microbiome in autoimmune diseases in HIV patients.
Collapse
Affiliation(s)
- Chiara Pellicano
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | - Giorgia Leodori
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | | | - Antonietta Gigante
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| | - Edoardo Rosato
- Department of Translational and Precision Medicine-Scleroderma Unit, Sapienza University of Rome, 00185 Rome, Italy.
| |
Collapse
|
238
|
Wingfield B, Coleman S, McGinnity TM, Bjourson AJ. Robust Microbial Markers for Non-Invasive Inflammatory Bowel Disease Identification. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:2078-2088. [PMID: 29994028 DOI: 10.1109/tcbb.2018.2831212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inflammatory Bowel Disease (IBD) is an umbrella term for a group of inflammatory diseases of the gastrointestinal tract, including Crohn's Disease and ulcerative colitis. Changes to the intestinal microbiome, the community of micro-organisms that resides in the human gut, have been shown to contribute to the pathogenesis of IBD. IBD diagnosis is often delayed due to its non-specific symptoms and because an invasive colonoscopy is required for confirmation, which leads to poor growth in children and worse treatment outcomes. Feature selection algorithms are often applied to microbial communities to identify bacterial groups that drive disease. It has been shown that aggregating Ensemble Feature Selection (EFS) can improve the robustness of feature selection algorithms, which is defined as the variation of feature selector output caused by small changes to the dataset. In this work, we apply a two-step filter and an EFS process to generate robust feature subsets that can non-invasively predict IBD subtypes from high-resolution microbiome data. The predictive power of the robust feature subsets is the highest reported in literature to date. Furthermore, we identify five biologically plausible bacterial species that have not previously been implicated in IBD aetiology.
Collapse
|
239
|
Fernández-Tomé S, Marin AC, Ortega Moreno L, Baldan-Martin M, Mora-Gutiérrez I, Lanas-Gimeno A, Moreno-Monteagudo JA, Santander C, Sánchez B, Chaparro M, Gisbert JP, Bernardo D. Immunomodulatory Effect of Gut Microbiota-Derived Bioactive Peptides on Human Immune System from Healthy Controls and Patients with Inflammatory Bowel Disease. Nutrients 2019; 11:nu11112605. [PMID: 31683517 PMCID: PMC6893616 DOI: 10.3390/nu11112605] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/18/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023] Open
Abstract
Bioactive peptides secreted by probiotic Bifidobacterium longum (peptide B7) and opportunistic pathogen Bacteroides fragilis (peptide B12) modulate the intestinal cytokine milieu in health. Here, we characterized their capacity to modulate both the mucosal cytokine production and the phenotype of circulating antigen presenting cells (APCs) in active inflammatory bowel disease (IBD). The IBD mucosa produced higher levels of pro-inflammatory cytokines referred to healthy controls (HCs). Peptides B7 and B12, however, did not ameliorate the mucosal cytokine milieu in IBD. Human circulating APCs (B-cells, monocytes, plasmacytoid dendritic cells (pDCs), and conventional dendritic cells (cDCs)) were characterized by flow cytometry in presence/absence of the peptides. Circulating B-cells, monocytes, and cDCs from IBD patients were more activated than those from HCs. Peptide B7, but not B12, decreased CCR2 expression on all APC subsets from HC, but not IBD patients. Moreover, both peptides tend to further increase their pro-inflammatory profile in IBD. In summary, IBD patients display mucosal and circulating APC pro-inflammatory properties. Peptide B7 immunomodulatory capacity elicited over circulating APCs from HC, but not IBD patients, suggests the presence of disrupted modulatory mechanisms for this peptide in IBD. Future studies should address the effect of bacteria-derived immunomodulatory peptides in non-inflamed (quiescent) IBD patients.
Collapse
Affiliation(s)
- Samuel Fernández-Tomé
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
| | - Alicia C Marin
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain.
| | - Lorena Ortega Moreno
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
| | - Montserrat Baldan-Martin
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain.
| | - Irene Mora-Gutiérrez
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
| | - Aitor Lanas-Gimeno
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
| | - José Andrés Moreno-Monteagudo
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
| | - Cecilio Santander
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
| | - Borja Sánchez
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Asturias, Spain.
| | - María Chaparro
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain.
| | - Javier P Gisbert
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain.
| | - David Bernardo
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain.
- Mucosal Immunology Lab, Instituto de Biología y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), 47003 Valladolid, Spain.
| |
Collapse
|
240
|
Bullich C, Keshavarzian A, Garssen J, Kraneveld A, Perez-Pardo P. Gut Vibes in Parkinson's Disease: The Microbiota-Gut-Brain Axis. Mov Disord Clin Pract 2019; 6:639-651. [PMID: 31745471 DOI: 10.1002/mdc3.12840] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/08/2019] [Accepted: 08/20/2019] [Indexed: 12/17/2022] Open
Abstract
Background The complexity of the pathogenic mechanisms underlying neurodegenerative disorders such as Parkinson's disease (PD) is attributable to multifactorial changes occurring at a molecular level, influenced by genetics and environmental interactions. However, what causes the main hallmarks of PD is not well understood. Recent data increasingly suggest that imbalances in the gut microbiome composition might trigger and/or exacerbate the progression of PD. Objective The present review aims to (1) report emerging literature showing changes in microbiota composition of PD patients compared to healthy individuals and (2) discuss how these changes may initiate and/or perpetuate PD pathology. Methods We analyzed 13 studies published from 2015 and included in this review. Altered microbial taxa were compiled in a detailed table summarizing bacterial changes in fecal/mucosal samples. The methodology was systematically reviewed across the articles and was also included in a table to facilitate comparisons between studies. Results Multiple studies found a reduction in short-chain fatty-acid-producing bacteria that can rescue neuronal damage through epigenetic mechanisms. Overall, the studies showed that changes in the gut microbiota composition might influence colonic inflammation, gut permeability, and α-synuclein aggregation, contributing to the neurogenerative process. Conclusion Further studies with larger cohorts and high-resolution sequencing methods are required to better define gut microbiota changes in PD. Furthermore, additional longitudinal studies are required to determine the causal link between these changes and PD pathogenesis as well as to study the potential of the intestinal microbiota as a biomarker.
Collapse
Affiliation(s)
- Clara Bullich
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands
| | - Ali Keshavarzian
- Department of Medicine, Division of Allergy-Immunology Rush University Medical Center Chicago Illinois USA
| | - Johan Garssen
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands.,Nutricia Reasearch Utrecht The Netherlands
| | - Aletta Kraneveld
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands.,Institute for Risk Assessment Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology Utrecht Institute for Pharmaceutical Sciences, Faculty of Science Utrecht University Utrecht The Netherlands
| |
Collapse
|
241
|
Yue B, Luo X, Yu Z, Mani S, Wang Z, Dou W. Inflammatory Bowel Disease: A Potential Result from the Collusion between Gut Microbiota and Mucosal Immune System. Microorganisms 2019; 7:microorganisms7100440. [PMID: 31614539 PMCID: PMC6843348 DOI: 10.3390/microorganisms7100440] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Host health depends on the intestinal homeostasis between the innate/adaptive immune system and the microbiome. Numerous studies suggest that gut microbiota are constantly monitored by the host mucosal immune system, and any slight disturbance in the microbial communities may contribute to intestinal immune disruption and increased susceptibility to inflammatory bowel disease (IBD), a chronic relapsing inflammatory condition of the gastrointestinal tract. Therefore, maintaining intestinal immune homeostasis between microbiota composition and the mucosal immune system is an effective approach to prevent and control IBD. The overall theme of this review is to summarize the research concerning the pathogenesis of IBD, with particular focus on the factors of gut microbiota-mucosal immune interactions in IBD. This is a comprehensive and in-depth report of the crosstalk between gut microbiota and the mucosal immune system in IBD pathogenesis, which may provide insight into the further evaluation of the therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Bei Yue
- Shanghai Key Laboratory of Formulated Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai 201203, China.
| | - Xiaoping Luo
- Shanghai Key Laboratory of Formulated Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai 201203, China.
| | - Zhilun Yu
- Shanghai Key Laboratory of Formulated Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai 201203, China.
| | - Sridhar Mani
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, The Bronx, NY 10461, USA.
| | - Zhengtao Wang
- Shanghai Key Laboratory of Formulated Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai 201203, China.
| | - Wei Dou
- Shanghai Key Laboratory of Formulated Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai 201203, China.
| |
Collapse
|
242
|
Singh V, Yeoh BS, Walker RE, Xiao X, Saha P, Golonka RM, Cai J, Bretin ACA, Cheng X, Liu Q, Flythe MD, Chassaing B, Shearer GC, Patterson AD, Gewirtz AT, Vijay-Kumar M. Microbiota fermentation-NLRP3 axis shapes the impact of dietary fibres on intestinal inflammation. Gut 2019; 68:1801-1812. [PMID: 30670576 DOI: 10.1136/gutjnl-2018-316250] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 12/06/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Diets rich in fermentable fibres provide an array of health benefits; however, many patients with IBD report poor tolerance to fermentable fibre-rich foods. Intervention studies with dietary fibres in murine models of colonic inflammation have yielded conflicting results on whether fibres ameliorate or exacerbate IBD. Herein, we examined how replacing the insoluble fibre, cellulose, with the fermentable fibres, inulin or pectin, impacted murine colitis resulting from immune dysregulation via inhibition of interleukin (IL)-10 signalling and/or innate immune deficiency (Tlr5KO). DESIGN Mice were fed with diet containing either cellulose, inulin or pectin and subjected to weekly injections of an IL-10 receptor (αIL-10R) neutralising antibody. Colitis development was examined by serological, biochemical, histological and immunological parameters. RESULTS Inulin potentiated the severity of αIL10R-induced colitis, while pectin ameliorated the disease. Such exacerbation of colitis following inulin feeding was associated with enrichment of butyrate-producing bacteria and elevated levels of caecal butyrate. Blockade of butyrate production by either metronidazole or hops β-acids ameliorated colitis severity in inulin-fed mice, whereas augmenting caecal butyrate via tributyrin increased colitis severity in cellulose containing diet-fed mice. Elevated butyrate levels were associated with increased IL-1β activity, while inhibition of the NOD-like receptor protein 3 by genetic, pharmacologic or dietary means markedly reduced colitis. CONCLUSION These results not only support the notion that fermentable fibres have the potential to ameliorate colitis but also caution that, in some contexts, prebiotic fibres can lead to gut dysbiosis and surfeit colonic butyrate that might exacerbate IBD.
Collapse
Affiliation(s)
- Vishal Singh
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Beng San Yeoh
- Nutritional Sciences, Graduate Program in Immunology and Infectious Diseases, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Rachel E Walker
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Xia Xiao
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Piu Saha
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Rachel M Golonka
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Jingwei Cai
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Alexis Charles Andre Bretin
- Center for Inflammation Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Xi Cheng
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Qing Liu
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Michael D Flythe
- USDA-Agriculture Research Service, University of Kentucky Campus, Lexington, Kentucky, USA
| | - Benoit Chassaing
- Center for Inflammation Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA.,Neuroscience Institute, Institutefor Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Gregory C Shearer
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Andrew T Gewirtz
- Center for Inflammation Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Matam Vijay-Kumar
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA.,Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
243
|
Klaassen MAY, Imhann F, Collij V, Fu J, Wijmenga C, Zhernakova A, Dijkstra G, Festen EAM, Gacesa R, Vich Vila A, Weersma RK. Anti-inflammatory Gut Microbial Pathways Are Decreased During Crohn's Disease Exacerbations. J Crohns Colitis 2019; 13:1439-1449. [PMID: 31066440 PMCID: PMC7142399 DOI: 10.1093/ecco-jcc/jjz077] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Crohn's disease [CD] is a chronic inflammatory disorder of the gastrointestinal tract characterised by alternating periods of exacerbation and remission. We hypothesised that changes in the gut microbiome are associated with CD exacerbations, and therefore aimed to correlate multiple gut microbiome features to CD disease activity. METHODS Faecal microbiome data generated using whole-genome metagenomic shotgun sequencing of 196 CD patients were of obtained from the 1000IBD cohort [one sample per patient]. Patient disease activity status at time of sampling was determined by re-assessing clinical records 3 years after faecal sample production. Faecal samples were designated as taken 'in an exacerbation' or 'in remission'. Samples taken 'in remission' were further categorised as 'before the next exacerbation' or 'after the last exacerbation', based on the exacerbation closest in time to the faecal production date. CD activity was correlated with gut microbial composition and predicted functional pathways via logistic regressions using MaAsLin software. RESULTS In total, 105 bacterial pathways were decreased during CD exacerbation (false-discovery rate [FDR] <0.1) in comparison with the gut microbiome of patients both before and after an exacerbation. Most of these decreased pathways exert anti-inflammatory properties facilitating the biosynthesis and fermentation of various amino acids [tryptophan, methionine, and arginine], vitamins [riboflavin and thiamine], and short-chain fatty acids [SCFAs]. CONCLUSIONS CD exacerbations are associated with a decrease in microbial genes involved in the biosynthesis of the anti-inflammatory mediators riboflavin, thiamine, and folate, and SCFAs, suggesting that increasing the intestinal abundances of these mediators might provide new treatment opportunities. These results were generated using bioinformatic analyses of cross-sectional data and need to be replicated using time-series and wet lab experiments.
Collapse
Affiliation(s)
- Marjolein A Y Klaassen
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Floris Imhann
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Valerie Collij
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Jingyuan Fu
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Cisca Wijmenga
- University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Alexandra Zhernakova
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Gerard Dijkstra
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Eleonora A M Festen
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Ranko Gacesa
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Arnau Vich Vila
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Rinse K Weersma
- University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands,University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands,Corresponding author: R. K. Weersma, MD, PhD, PO Box 30.001, 9700RB Groningen, The Netherlands. Tel.: +316 41132824; Fax 050 361 9306;
| |
Collapse
|
244
|
Niccolai E, Baldi S, Ricci F, Russo E, Nannini G, Menicatti M, Poli G, Taddei A, Bartolucci G, Calabrò AS, Stingo FC, Amedei A. Evaluation and comparison of short chain fatty acids composition in gut diseases. World J Gastroenterol 2019; 25:5543-5558. [PMID: 31576099 PMCID: PMC6767983 DOI: 10.3748/wjg.v25.i36.5543] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND An altered (dysbiosis) and unhealthy status of the gut microbiota is usually responsible for a reduction of short chain fatty acids (SCFAs) concentration. SCFAs obtained from the carbohydrate fermentation processes are crucial in maintaining gut homeostasis and their determination in stool samples could provide a faster, reliable and cheaper method to highlight the presence of an intestinal dysbiosis and a biomarker for various gut diseases. We hypothesize that different intestinal diseases, such as celiac disease (CD), adenomatous polyposis (AP) and colorectal cancer (CRC) could display a particular fecal SCFAs' signature. AIM To compare the fecal SCFAs' profiles of CD, AP, CRC patients and healthy controls, using the same analytical method. METHODS In this cross-sectional study, we defined and compared the SCFAs' concentration in fecal samples of 9 AP, 16 CD, 19 CRC patients and 16 healthy controls (HC). The SCFAs' analysis were performed using a gas-chromatography coupled with mass spectrometry method. Data analysis was carried out using Wilcoxon rank-sum test to assess pairwise differences of SCFAs' profiles, partial least squares-discriminate analysis (PLS-DA) to determine the status membership based on distinct SCFAs' profiles, and Dirichlet regression to determine factors influencing concentration levels of SCFAs. RESULTS We have not observed any difference in the SCFAs' amount and composition between CD and healthy control. On the contrary, the total amount of SCFAs was significantly lower in CRC patients compared to HC (P = 0.044) and CD (P = 0.005). Moreover, the SCFAs' percentage composition was different in CRC and AP compared to HC. In detail, HC displayed higher percentage of acetic acid (P value = 1.3 × 10-6) and a lower amount of butyric (P value = 0.02192), isobutyric (P value = 7.4 × 10-5), isovaleric (P value = 0.00012) and valeric (P value = 0.00014) acids compared to CRC patients. AP showed a lower abundance of acetic acid (P value = 0.00062) and higher percentages of propionic (P value = 0.00433) and isovaleric (P value = 0.00433) acids compared to HC. Moreover, AP showed higher levels of propionic acid (P value = 0.03251) and a lower level of isobutyric acid (P value = 0.00427) in comparison to CRC. The PLS-DA model demonstrated a significant separation of CRC and AP groups from HC, although some degree of overlap was observed between CRC and AP. CONCLUSION Analysis of fecal SCFAs shows the potential to provide a non-invasive means of diagnosis to detect patients with CRC and AP, while CD patients cannot be discriminated from healthy subjects.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Federica Ricci
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” University of Florence, Florence 50134, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Marta Menicatti
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, Florence 50134, Italy
| | - Giovanni Poli
- Department of Statistics, Computer Science, Applications “G.Parenti”, Florence 50134, Italy
| | - Antonio Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, Florence 50134, Italy
| | - Antonino Salvatore Calabrò
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” University of Florence, Florence 50134, Italy
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
- Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi, Florence 50134, Italy
| |
Collapse
|
245
|
Ungaro F, Massimino L, D'Alessio S, Danese S. The gut virome in inflammatory bowel disease pathogenesis: From metagenomics to novel therapeutic approaches. United European Gastroenterol J 2019; 7:999-1007. [PMID: 31662858 DOI: 10.1177/2050640619876787] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022] Open
Abstract
The association of intestinal dysbiosis with the pathogenesis of inflammatory bowel disease has been well established. Besides bacteria, microbiota comprises yeasts, archaea, protists and viruses, neglected actors in inflammatory bowel disease-associated microbiota. In the past, a great limitation in studying microbiota composition was the low sensitivity of sequencing technologies and that few computational approaches were sufficient to thoroughly analyse the whole microbiome. However, new cutting-edge technologies in nucleic acid sequencing, -omics analysis and the innovative statistics and bioinformatics pipelines made possible more sensitive and accurate metagenomics, ultimately identifying novel players in intestinal inflammation, including prokaryotic and eukaryotic viruses, that together form the gut virome. The discovery of peculiar inflammatory bowel disease-associated microbial strains will not only shed new light on inflammatory bowel disease aetiogenesis, they may also support the development of novel therapeutic strategies not merely treating symptoms, but precisely counteracting the primary cause of chronic intestinal inflammation.
Collapse
Affiliation(s)
- Federica Ungaro
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Luca Massimino
- Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Silvia D'Alessio
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Silvio Danese
- IBD Center, Laboratory of Gastrointestinal Immunopathology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
246
|
Wu M, Li J, An Y, Li P, Xiong W, Li J, Yan D, Wang M, Zhong G. Chitooligosaccharides Prevents the Development of Colitis-Associated Colorectal Cancer by Modulating the Intestinal Microbiota and Mycobiota. Front Microbiol 2019; 10:2101. [PMID: 31620100 PMCID: PMC6759605 DOI: 10.3389/fmicb.2019.02101] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 08/26/2019] [Indexed: 12/20/2022] Open
Abstract
Gut microbes play a crucial role in the development of colorectal cancer. Chitooligosaccharides (COS), are oligomer that are depolymerized from chitosan and possess a wide range of biological activities. In this study, the effects of COS on colorectal cancer (CRC) development were evaluated using azoxymethane and dextran sulfate sodium (AOM/DSS) induced mouse model of CRC (CACM). In the COS-treated CRC group (CMCOS), COS protected mice from CRC by decreasing the disease activity index, tumor incidences and multiplicity, and the mRNA levels of COX-2, IL-6, TNF-α, IL-1β, IL-10, and IKK-β mRNA in colonic epithelial cells. The results of a cage-exchanged experiment, in which mice from the CACMe and CMCOSe treatments exchanged cages every day to interact with microbes, showed that gut microbes play an important role in preventing CAC by COS. The abundances of fecal bacteria (total bacteria, Lactobacillus, Enterococcus, Fusobacterium nucleatum and butyrate-producing bacteria) were detected by qPCR on the 0th, 1st, 3rd, 6th, 9th, and 10th weekends. Furthermore, microbiota and mycobiota were analyzed by high-throughput sequencing on an Illumina MiSeq PE300 system. COS protected mice from CRC by reversing the imbalance of bacteria and fungi, especially by reducing the abundance of Escherichia-Shigella, Enterococcus, and Turicibacter, and increasing the levels of Akkermansia, butyrate-producing bacteria and Cladosporium.
Collapse
Affiliation(s)
- Minna Wu
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jianmin Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Shaanxi Provincial Second People’s Hospital, Xi’an, China
| | - Yunying An
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Puze Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Wancheng Xiong
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jinsong Li
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Dong Yan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Mingyong Wang
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Genshen Zhong
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
247
|
Adenovirus infection is associated with altered gut microbial communities in a non-human primate. Sci Rep 2019; 9:13410. [PMID: 31527752 PMCID: PMC6746978 DOI: 10.1038/s41598-019-49829-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
Adenovirus (AdV) infections are one of the main causes of diarrhea in young children. Enteric AdVs probably disrupt gut microbial defences, which can result in diarrhea. To understand the role of the gut microbiome in AdV-induced pathologies, we investigated the gut microbiome of a naturally AdV-infected non-human primate species, the Malagasy mouse lemur (Microcebus griseorufus), which represents an important model in understanding the evolution of diseases. We observed that AdV infection is associated with disruption of the gut microbial community composition. In AdV+ lemurs, several commensal taxa essential for a healthy gut microbiome decreased, whereas genera containing potential pathogens, such as Neisseria, increased in abundance. Microbial co-occurrence networks revealed a loss of important microbial community interactions in AdV+ lemurs and an overrepresentation of Prevotellaceae. The observation of enteric virus-associated loss of commensal bacteria and associated shifts towards pathobionts may represent the missing link for a better understanding of AdV-induced effects in humans, and also for their potential as drivers of co-infections, an area of research that has been largely neglected so far.
Collapse
|
248
|
Schepici G, Silvestro S, Bramanti P, Mazzon E. The Gut Microbiota in Multiple Sclerosis: An Overview of Clinical Trials. Cell Transplant 2019; 28:1507-1527. [PMID: 31512505 PMCID: PMC6923550 DOI: 10.1177/0963689719873890] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory, demyelinating, and
degenerative disease that affects the central nervous system. A recent study
showed that interaction between the immune system and the gut microbiota plays a
crucial role in the development of MS. This review reports the clinical studies
carried out in recent years that aimed to evaluate the composition of the
microbiota in patients with relapsing–remitting MS (RR-MS). We also report what
is available in the literature regarding the effectiveness of fecal microbiota
transplantation and the role of the diet in restoring the intestinal bacterial
population. Studies report that patients with RR-MS have a microbiota that,
compared with healthy controls, has higher amounts of
Pedobacteria, Flavobacterium,
Pseudomonas, Mycoplana,
Acinetobacter, Eggerthella,
Dorea, Blautia,
Streptococcus and Akkermansia. In
contrast, MS patients have a microbiota with impoverished microbial populations
of Prevotella, Bacteroides,
Parabacteroides, Haemophilus,
Sutterella, Adlercreutzia,
Coprobacillus, Lactobacillus,
Clostridium, Anaerostipes and
Faecalibacterium. In conclusion, the restoration of the
microbial population in patients with RR-MS appears to reduce inflammatory
events and the reactivation of the immune system.
Collapse
Affiliation(s)
- Giovanni Schepici
- IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy.,Both the authors contributed equally to this article
| | - Serena Silvestro
- IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy.,Both the authors contributed equally to this article
| | | | | |
Collapse
|
249
|
Galazzo G, Tedjo DI, Wintjens DSJ, Savelkoul PHM, Masclee AAM, Bodelier AGL, Pierik MJ, Jonkers DMAE, Penders J. Faecal Microbiota Dynamics and their Relation to Disease Course in Crohn's Disease. J Crohns Colitis 2019; 13:1273-1282. [PMID: 30810207 PMCID: PMC6764104 DOI: 10.1093/ecco-jcc/jjz049] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Microbial shifts have been associated with disease activity in Crohn's disease [CD], but findings on specific taxa are inconsistent. This may be due to differences in applied methods and cross-sectional study designs. We prospectively examined the faecal microbiota in adult CD patients with changing or stable disease course over time. METHODS Faeces were collected at two time-points from 15 healthy control individuals [HCs], 35 CD patients who were in remission and who maintained remission [RRs], and 22 CD patients during remission and also during subsequent exacerbation [RAs]. The microbial composition was assessed by 16S rRNA [V4] gene sequencing. RESULTS Compared with HCs, patients with CD had a lower microbial richness [p = 0.0002] and diversity [p = 0.005]. Moreover, the microbial community structure of a subset of patients, clustered apart from HCs, was characterized by low microbial diversity and Faecalibacterium abundance. Patients within this cluster did not differ with respect to long-term disease course compared with patients with a 'healthy-appearing' microbiota.Over time, microbial richness and diversity did not change in RR versus RA patients. Although the microbial community structure of both RR and RA patients was less stable over time compared with that of HCs, no differences were observed between the patient groups [p = 0.17]; nor was the stability impacted by Montreal classification, medication use, or surgery. CONCLUSION The altered microbiota composition and stability in CD was neither associated with disease activity nor long-term disease course, questioning its involvement in the development of an exacerbation. The aberrant microbiota composition in a subset of CD patients warrants further exploration of a more microbiota-driven etiology in this group.
Collapse
Affiliation(s)
- Gianluca Galazzo
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands,School of Public Health and Primary Care [Caphri], Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Danyta I Tedjo
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands,School of Nutrition and Translational Research in Metabolism [NUTRIM], Division Gastroenterology–Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Dion S J Wintjens
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Division Gastroenterology–Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Paul H M Savelkoul
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands,School of Public Health and Primary Care [Caphri], Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands,Department of Medical Microbiology & Infection control, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ad A M Masclee
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Division Gastroenterology–Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Marie J Pierik
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Division Gastroenterology–Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Daisy M A E Jonkers
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Division Gastroenterology–Hepatology, Maastricht University Medical Center+, Maastricht, The Netherlands,Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - John Penders
- School of Nutrition and Translational Research in Metabolism [NUTRIM], Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands,School of Public Health and Primary Care [Caphri], Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands,Corresponding authors: J. Penders, Department of Medical Microbiology, Maastricht University Medical Center+, PO 5800, 6202 AZ, Maastricht, The Netherlands. Tel: +31-(0)433875134; Fax: +31-(0)433676643;
| |
Collapse
|
250
|
Han MM, Zhu XY, Peng YF, Lin H, Liu DC, Li L. The alterations of gut microbiota in mice with chronic pancreatitis. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:464. [PMID: 31700900 DOI: 10.21037/atm.2019.08.18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The changes of intestinal microbiome are associated with inflammatory, metabolic, and malignant disorders, and there are no studies assessing the intestinal microbiota of mice with chronic pancreatitis (CP). Thus, we aim to investigate the variations in diversity, composition and function of intestinal microbiota in CP mice. Methods Sixteen male C57BL/6 mice were randomly selected, and divided into two groups, treated intraperitoneally with saline (normal control group, CT group) or ethanol + cerulein (experimental group, CP group) for 6 weeks. Body weight as measured in entire processes. Histopathological examination of CP index was conducted to verify the CP induction. Extracted DNA from colon samples was used for Illumina HiSeq sequencing of the bacterial V4 region of 16S rRNA gene and analyzed using Quantitative Insights Into Microbial Ecology (QIIME). Functional profiling of microbial communities was predicted with BugBase. Results Significant alterations of the gut microbiota were found in the CP mice compared to CT groups, as revealed by significant decrease in bacterial richness and diversity, declined the relative abundance of Lachnospiraceae_NK4A136, Ruminiclostridium and Roseburia, and increased the relative abundances of Bacteroides and Alloprevotella genera. Analysis of microbial community-level phenotypes revealed significant differences in nine phenotypes (aerobic, anaerobic, containing mobile elements, facultatively anaerobic, biofilm forming, gram-negative, gram-positive, potentially pathogenic, and stress tolerant) between CP group and CT group. Conclusions This study indicated that mice with CP had a distinct microbiota profile.
Collapse
Affiliation(s)
- Man-Man Han
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.,Pancreatic Research Institute, Southeast University, Nanjing 210009, China
| | - Xiang-Yun Zhu
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.,Pancreatic Research Institute, Southeast University, Nanjing 210009, China
| | - You-Fan Peng
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.,Pancreatic Research Institute, Southeast University, Nanjing 210009, China
| | - Hao Lin
- Pancreatic Research Institute, Southeast University, Nanjing 210009, China.,Department of Clinical Science and Research, School of Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - De-Chen Liu
- Pancreatic Research Institute, Southeast University, Nanjing 210009, China.,Department of Clinical Science and Research, School of Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.,Pancreatic Research Institute, Southeast University, Nanjing 210009, China
| |
Collapse
|