101
|
Abstract
The diet and gut microbiota have been extensively interrogated as a fuel for gut inflammation in inflammatory bowel diseases (IBDs) in the last few years. Here, we review how specific nutrients, typically enriched in a Western diet, instigate or deteriorate experimental gut inflammation in a genetically susceptible host and we discuss microbiota-dependent and independent mechanisms. We depict the study landscape of nutritional trials in paediatric and adult IBD and delineate common grounds for dietary advice. Conclusively, the diet reflects a critical rheostat of microbial dysbiosis and gut inflammation in IBD. Dietary restriction by exclusive enteral nutrition, with or without a specific exclusion diet, is effectively treating paediatric Crohn's disease, while adult IBD trials are less conclusive. Insights into molecular mechanisms of nutritional therapy will change the perception of IBD and will allow us to enter the era of precision nutrition. To achieve this, we discuss the need for carefully designed nutritional trials with scientific rigour comparable to medical trials, which also requires action from stake holders. Establishing evidence-based dietary therapy for IBD does not only hold promise to avoid long-term immunosuppression, but to provide a widely accessible therapy at low cost. Identification of dietary culprits disturbing gut health also bears the potential to prevent IBD and allows informed decision making in food politics.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Medicine I, Gastroenterology, Hepatology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Jingwan Zhang
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
102
|
Holle J, Bartolomaeus H, Löber U, Behrens F, Bartolomaeus TU, Anandakumar H, Wimmer MI, Vu DL, Kuhring M, Brüning U, Maifeld A, Geisberger S, Kempa S, Schumacher F, Kleuser B, Bufler P, Querfeld U, Kitschke S, Engler D, Kuhrt LD, Drechsel O, Eckardt KU, Forslund SK, Thürmer A, McParland V, Kirwan JA, Wilck N, Müller D. Inflammation in Children with CKD Linked to Gut Dysbiosis and Metabolite Imbalance. J Am Soc Nephrol 2022; 33:2259-2275. [PMID: 35985814 PMCID: PMC9731629 DOI: 10.1681/asn.2022030378] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/29/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND CKD is characterized by a sustained proinflammatory response of the immune system, promoting hypertension and cardiovascular disease. The underlying mechanisms are incompletely understood but may be linked to gut dysbiosis. Dysbiosis has been described in adults with CKD; however, comorbidities limit CKD-specific conclusions. METHODS We analyzed the fecal microbiome, metabolites, and immune phenotypes in 48 children (with normal kidney function, CKD stage G3-G4, G5 treated by hemodialysis [HD], or kidney transplantation) with a mean±SD age of 10.6±3.8 years. RESULTS Serum TNF-α and sCD14 were stage-dependently elevated, indicating inflammation, gut barrier dysfunction, and endotoxemia. We observed compositional and functional alterations of the microbiome, including diminished production of short-chain fatty acids. Plasma metabolite analysis revealed a stage-dependent increase of tryptophan metabolites of bacterial origin. Serum from patients on HD activated the aryl hydrocarbon receptor and stimulated TNF-α production in monocytes, corresponding to a proinflammatory shift from classic to nonclassic and intermediate monocytes. Unsupervised analysis of T cells revealed a loss of mucosa-associated invariant T (MAIT) cells and regulatory T cell subtypes in patients on HD. CONCLUSIONS Gut barrier dysfunction and microbial metabolite imbalance apparently mediate the proinflammatory immune phenotype, thereby driving the susceptibility to cardiovascular disease. The data highlight the importance of the microbiota-immune axis in CKD, irrespective of confounding comorbidities.
Collapse
Affiliation(s)
- Johannes Holle
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Hendrik Bartolomaeus
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Behrens
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Institute of Physiology, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Theda U.P. Bartolomaeus
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Harithaa Anandakumar
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz I. Wimmer
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine IV, University Hospital of Tübingen, Tübingen, Germany
| | - Dai Long Vu
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Core Unit Metabolomics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Kuhring
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Ulrike Brüning
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Core Unit Metabolomics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Andras Maifeld
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sabrina Geisberger
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- The Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- The Berlin Institute for Medical Systems Biology, Berlin, Germany
| | | | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Querfeld
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Stefanie Kitschke
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Denise Engler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Leonard D. Kuhrt
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | | | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sofia K. Forslund
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Andrea Thürmer
- MF2 Genome Sequencing, Robert Koch Institute, Berlin, Germany
| | - Victoria McParland
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jennifer A. Kirwan
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Core Unit Metabolomics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center, a cooperation of Charité–Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Dominik Müller
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
103
|
Dubik M, Pilecki B, Moeller JB. Commensal Intestinal Protozoa-Underestimated Members of the Gut Microbial Community. BIOLOGY 2022; 11:1742. [PMID: 36552252 PMCID: PMC9774987 DOI: 10.3390/biology11121742] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
The human gastrointestinal microbiota contains a diverse consortium of microbes, including bacteria, protozoa, viruses, and fungi. Through millennia of co-evolution, the host-microbiota interactions have shaped the immune system to both tolerate and maintain the symbiotic relationship with commensal microbiota, while exerting protective responses against invading pathogens. Microbiome research is dominated by studies describing the impact of prokaryotic bacteria on gut immunity with a limited understanding of their relationship with other integral microbiota constituents. However, converging evidence shows that eukaryotic organisms, such as commensal protozoa, can play an important role in modulating intestinal immune responses as well as influencing the overall health of the host. The presence of several protozoa species has recently been shown to be a common occurrence in healthy populations worldwide, suggesting that many of these are commensals rather than invading pathogens. This review aims to discuss the most recent, conflicting findings regarding the role of intestinal protozoa in gut homeostasis, interactions between intestinal protozoa and the bacterial microbiota, as well as potential immunological consequences of protozoa colonization.
Collapse
Affiliation(s)
- Magdalena Dubik
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Bartosz Pilecki
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Jesper Bonnet Moeller
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Danish Institute for Advanced Study, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
104
|
Buffet-Bataillon S, Bouguen G, Fleury F, Cattoir V, Le Cunff Y. Gut microbiota analysis for prediction of clinical relapse in Crohn's disease. Sci Rep 2022; 12:19929. [PMID: 36402792 PMCID: PMC9675750 DOI: 10.1038/s41598-022-23757-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 11/04/2022] [Indexed: 11/20/2022] Open
Abstract
The role of intestinal bacterial microbiota has been described as key in the pathophysiology of Crohn's disease (CD). CD is characterized by frequent relapses after periods of remission which are not entirely understood. In this paper, we investigate whether the heterogeneity in microbiota profiles in CD patients could be a suitable predictor for these relapses. This prospective observational study involved 259 CD patients, in which 41 provided an additional total of 62 consecutive fecal samples, with an average interval of 25 weeks in between each of these samples. Fecal microbiota was analyzed by massive genomic sequencing through 16 S rRNA amplicon sampling. We found that our 259 CD patients could be split into three distinct subgroups of microbiota (G1, G2, G3). From G1 to G3, we noticed a progressive decrease in alpha diversity (p ≤ 0.0001) but no change in the fecal calprotectin (FC) level. Focusing on the 103 consecutive samples from 41 CD patients, we showed that the patients microbiota profiles were remarkably stable over time and associated with increasing symptom severity. Investigating further this microbiota/severity association revealed that the first signs of aggravation are (1) a loss of the main anti-inflammatory Short-Chain Fatty Acids (SCFAs) Roseburia, Eubacterium, Subdoligranumum, Ruminococcus (P < 0.05), (2) an increase in pro-inflammatory pathogens Proteus, Finegoldia (P < 0.05) while (3) an increase of other minor SCFA producers such as Ezakiella, Anaerococcus, Megasphaera, Anaeroglobus, Fenollaria (P < 0.05). Further aggravation of clinical signs is significantly linked to the subsequent loss of these minor SCFAs species and to an increase in other proinflammatory Proteobacteria such as Klebsiella, Pseudomonas, Salmonella, Acinetobacter, Hafnia and proinflammatory Firmicutes such as Staphylococcus, Enterococcus, Streptococcus. (P < 0.05). To our knowledge, this is the first study (1) specifically identifying subgroups of microbiota profiles in CD patients, (2) relating these groups to the evolution of symptoms over time and (3) showing a two-step process in CD symptoms' worsening. This paves the way towards a better understanding of patient-to-patient heterogeneity, as well as providing early warning signals of future aggravation of the symptoms and eventually adapting empirically treatments.
Collapse
Affiliation(s)
- Sylvie Buffet-Bataillon
- grid.410368.80000 0001 2191 9284INSERM, Institut NUMECAN (Nutrition Metabolisms and Cancer), CHU Rennes, Université Rennes 1, 35000 Rennes, France
| | - Guillaume Bouguen
- grid.410368.80000 0001 2191 9284CIC 1414, INSERM, Institut NUMECAN (Nutrition Metabolisms and Cancer), CHU Rennes, Université Rennes 1, 35000 Rennes, France
| | - François Fleury
- grid.410368.80000 0001 2191 9284INSERM, Institut NUMECAN (Nutrition Metabolisms and Cancer), CHU Rennes, Université Rennes 1, 35000 Rennes, France
| | - Vincent Cattoir
- grid.410368.80000 0001 2191 9284U1230, INSERM, CHU Rennes, Université Rennes 1, 35000 Rennes, France
| | - Yann Le Cunff
- grid.410368.80000 0001 2191 9284Dyliss - Dynamics, Logics and Inference for biological Systems and Sequences, Inria Rennes – Bretagne Atlantique, Université Rennes 1, Rennes, France
| |
Collapse
|
105
|
Li J, Ma Y, Li X, Wang Y, Huo Z, Lin Y, Li J, Yang H, Zhang Z, Yang P, Zhang C. Fermented Astragalus and its metabolites regulate inflammatory status and gut microbiota to repair intestinal barrier damage in dextran sulfate sodium-induced ulcerative colitis. Front Nutr 2022; 9:1035912. [PMID: 36451737 PMCID: PMC9702530 DOI: 10.3389/fnut.2022.1035912] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/27/2022] [Indexed: 09/20/2023] Open
Abstract
Fermentation represents an efficient biotechnological approach to increase the nutritional and functional potential of traditional Chinese medicine. In this study, Lactobacillus plantarum was used to ferment traditional Chinese medicine Astragalus, the differential metabolites in the fermented Astragalus (FA) were identified by ultra-performance liquid chromatography-Q Exactive hybrid quadrupole-Orbitrap mass spectrometry (UPLC-Q-Exactive-MS), and the ameliorating effect of FA on dextran sulfate sodium (DSS)-induced colitis in mice were further explored. The results showed that 11 differential metabolites such as raffinose, progesterone and uridine were identified in FA, which may help improve the ability of FA to alleviate colitis. Prophylactic FA supplementation effectively improved DAI score, colon length and histopathological lesion in DSS-treated mice. The abnormal activation of the intestinal immune barrier in mice was controlled after FA supplementation, the contents of myeloperoxidase (MPO) and IgE were reduced and the contents of IgA were increased. The intestinal pro-inflammatory factors TNF-α, IL-1β, IL-6, and IL-17 were down-regulated and the anti-inflammatory factors IL-10 and TGF-β were up-regulated, suggesting that FA can intervene in inflammatory status by regulating the balance of Th1/Th2/Th17/Treg related cytokines. In addition, FA supplementation modified the structure of the intestinal microbiota and enriched the abundance of Akkermansia and Alistipes, which were positively associated with the production of short-chain fatty acids. These microbes and their metabolites induced by FA also be involved in maintaining the intestinal mucosal barrier integrity by affecting mucosal immunity. We observed that intestinal tight junction protein and mucous secreting protein ZO-1, occludin, and MUC2 genes expression were more pronounced in mice supplemented with FA compared to unfermented Astragalus, along with modulation of intestinal epithelial cells (IECs) apoptosis, verifying the intestinal mucosal barrier repaired by FA. This study is the first to suggest that FA as a potential modulator can more effectively regulate the inflammatory status and gut microbiota to repair the intestinal barrier damage caused by colitis.
Collapse
Affiliation(s)
- Junxiang Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yingchun Ma
- Gansu Institute for Drug Control, Lanzhou, China
| | - Xiaofeng Li
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Yafei Wang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zeqi Huo
- School of Life Sciences, Lanzhou University, Lanzhou, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Yang Lin
- School of Life Sciences, Lanzhou University, Lanzhou, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Jiaru Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hui Yang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zhiming Zhang
- Gansu Provincial Hospital of Traditional Chinese Medicine (TCM), Lanzhou, China
| | - Pingrong Yang
- School of Pharmacy, Lanzhou University, Lanzhou, China
- Gansu Institute for Drug Control, Lanzhou, China
| | - Chunjiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| |
Collapse
|
106
|
Leibovitzh H, Lee SH, Xue M, Raygoza Garay JA, Hernandez-Rocha C, Madsen KL, Meddings JB, Guttman DS, Espin-Garcia O, Smith MI, Goethel A, Griffiths AM, Moayyedi P, Steinhart AH, Panaccione R, Huynh HQ, Jacobson K, Aumais G, Mack DR, Abreu MT, Bernstein CN, Marshall JK, Turner D, Xu W, Turpin W, Croitoru K. Altered Gut Microbiome Composition and Function Are Associated With Gut Barrier Dysfunction in Healthy Relatives of Patients With Crohn's Disease. Gastroenterology 2022; 163:1364-1376.e10. [PMID: 35850197 DOI: 10.1053/j.gastro.2022.07.004] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The gut microbiome has been suggested to play a role in gut barrier hemostasis, but data are scarce and limited to animal studies. We therefore aimed to assess whether alterations in gut microbial composition and functional pathways are associated with gut barrier function in a cohort of healthy first-degree relatives of patients with Crohn's disease. METHODS We used the Crohn's and Colitis Canada Genetic Environmental Microbial (CCC-GEM) cohort of healthy first-degree relatives of patients with Crohn's disease. Gut barrier function was assessed using the urinary fractional excretion of lactulose-to-mannitol ratio (LMR). Microbiome composition was assessed by sequencing fecal 16S ribosomal RNA. The cohort was divided into a discovery cohort (n = 2472) and a validation cohort (n = 655). A regression model was used to assess microbial associations with the LMR. A random forest classifier algorithm was performed to assess microbial community contribution to barrier function. RESULTS Individuals with impaired barrier function (LMR >0.025) had reduced alpha-diversity (Chao1 index, P = 4.0e-4) and altered beta-diversity (Bray-Curtis dissimilarity index, R2 = 0.001, P = 1.0e-3) compared with individuals with an LMR ≤0.025. When taxa were assessed individually, we identified 8 genera and 52 microbial pathways associated with an LMR >0.025 (q < 0.05). Four genera (decreased prevalence of Adlercreutzia, Clostridia UCG 014, and Clostridium sensu stricto 1 and increased abundance of Colidextribacter) and 8 pathways (including decreased biosynthesis of glutamate, tryptophan, and threonine) were replicated in the validation cohort. The random forest approach revealed that the bacterial community is associated with gut barrier function (area under the curve, 0.63; P = 1.4e-6). CONCLUSIONS The gut microbiome community and pathways are associated with changes in gut barrier function. These findings may identify potential microbial targets to modulate gut barrier.
Collapse
Affiliation(s)
- Haim Leibovitzh
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sun-Ho Lee
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyue Xue
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Juan Antonio Raygoza Garay
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Cristian Hernandez-Rocha
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Jonathan B Meddings
- Department of Medicine, Cumming School of Medicine, Calgary, Alberta, Canada
| | - David S Guttman
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada; Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Osvaldo Espin-Garcia
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Michelle I Smith
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ashleigh Goethel
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anne M Griffiths
- IBD Center, The Hospital for Sick Children, Department of Paediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Paul Moayyedi
- Department of Medicine, McMaster University, Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| | - A Hillary Steinhart
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Remo Panaccione
- Inflammatory Bowel Disease Clinic, Division of Gastroenterology and Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Hien Q Huynh
- Division of Gastroenterology and Nutrition, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Kevan Jacobson
- Canadian Gastro-Intestinal Epidemiology Consortium, Toronto, Ontario, Canada; British Columbia Children's Hospital, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Guy Aumais
- Department of Medicine, Hôpital Maisonneuve-Rosemont, Montreal University, Montreal, Quebec, Canada
| | - David R Mack
- Division of Gastroenterology, Hepatology & Nutrition, Children's Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
| | - Maria T Abreu
- Division of Gastroenterology, Crohn's and Colitis Center, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Charles N Bernstein
- University of Manitoba Inflammatory Bowel Disease Clinical and Research Centre and Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - John K Marshall
- Department of Medicine, McMaster University, Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| | - Dan Turner
- The Juliet Keidan Institute of Pediatric Gastroenterology and Nutrition, Shaare Zedek Medical Center, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wei Xu
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | | | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
107
|
Lekang K, Shekhar S, Berild D, Petersen FC, Winther-Larsen HC. Effects of different amoxicillin treatment durations on microbiome diversity and composition in the gut. PLoS One 2022; 17:e0275737. [PMID: 36301847 PMCID: PMC9612567 DOI: 10.1371/journal.pone.0275737] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 09/22/2022] [Indexed: 11/05/2022] Open
Abstract
Antibiotics seize an effect on bacterial composition and diversity and have been demonstrated to induce disruptions on gut microbiomes. This may have implications for human health and wellbeing, and an increasing number of studies suggest a link between the gut microbiome and several diseases. Hence, reducing antibiotic treatments may be beneficial for human health status. Further, antimicrobial resistance (AMR) is an increasing global problem that can be counteracted by limiting the usage of antibiotics. Longer antibiotic treatments have been demonstrated to increase the development of AMR. Therefore, shortening of antibiotic treatment durations, provided it is safe for patients, may be one measure to reduce AMR. In this study, the objective was to investigate effects of standard and reduced antibiotic treatment lengths on gut microbiomes using a murine model. Changes in the murine gut microbiome was assessed after using three different treatment durations of amoxicillin (3, 7 or 14 days) as well as a control group not receiving amoxicillin. Fecal samples were collected before and during the whole experiment, until three weeks past end of treatment. These were further subject for 16S rRNA Illumina MiSeq sequencing. Our results demonstrated significant changes in bacterial diversity, richness and evenness during amoxicillin treatment, followed by a reversion in terms of alpha-diversity and abundance of major phyla, after end of treatment. However, a longer restitution time was indicated for mice receiving amoxicillin for 14 days, and phylum Patescibacteria did not fully recover. In addition, an effect on the composition of Firmicutes was indicated to last for at least three weeks in mice treated with amoxicillin for 14 days. Despite an apparently reversion to a close to original state in overall bacterial diversity and richness, the results suggested more durable changes in lower taxonomical levels. We detected several families, genera and ASVs with significantly altered abundance three weeks after exposure to amoxicillin, as well as bacterial taxa that appeared significantly affected by amoxicillin treatment length. This may strengthen the argument for shorter antibiotic treatment regimens to both limit the emergence of antibiotic resistance and risk of gut microbiome disturbance.
Collapse
Affiliation(s)
- Katrine Lekang
- Department of Pharmacy, Section for Pharmacology and Pharmaceutical Biosciences, University of Oslo, Oslo, Norway
| | - Sudhanshu Shekhar
- Faculty of Dentistry, Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Dag Berild
- Faculty of Medicine, Department of Infectious Diseases, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Hanne C. Winther-Larsen
- Department of Pharmacy, Section for Pharmacology and Pharmaceutical Biosciences, University of Oslo, Oslo, Norway
- * E-mail:
| |
Collapse
|
108
|
Metabolic Modeling and Bidirectional Culturing of Two Gut Microbes Reveal Cross-Feeding Interactions and Protective Effects on Intestinal Cells. mSystems 2022; 7:e0064622. [PMID: 36005398 PMCID: PMC9600892 DOI: 10.1128/msystems.00646-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota is constituted by thousands of microbial interactions, some of which correspond to the exchange of metabolic by-products or cross-feeding. Inulin and xylan are two major dietary polysaccharides that are fermented by members of the human gut microbiota, resulting in different metabolic profiles. Here, we integrated community modeling and bidirectional culturing assays to study the metabolic interactions between two gut microbes, Phocaeicola dorei and Lachnoclostridium symbiosum, growing in inulin or xylan, and how they provide a protective effect in cultured cells. P. dorei (previously belonging to the Bacteroides genus) was able to consume inulin and xylan, while L. symposium only used certain inulin fractions to produce butyrate as a major end product. Constrained-based flux simulations of refined genome-scale metabolic models of both microbes predicted high lactate and succinate cross-feeding fluxes between P. dorei and L. symbiosum when growing in each fiber. Bidirectional culture assays in both substrates revealed that L. symbiosum growth increased in the presence of P. dorei. Carbohydrate consumption analyses showed a faster carbohydrate consumption in cocultures compared to monocultures. Lactate and succinate concentrations in bidirectional cocultures were lower than in monocultures, pointing to cross-feeding as initially suggested by the model. Butyrate concentrations were similar across all conditions. Finally, supernatants from both bacteria cultured in xylan in bioreactors significantly reduced tumor necrosis factor-α-induced inflammation in HT-29 cells and exerted a protective effect against the TcdB toxin in Caco-2 epithelial cells. Surprisingly, this effect was not observed in inulin cocultures. Overall, these results highlight the predictive value of metabolic models integrated with microbial culture assays for probing microbial interactions in the gut microbiota. They also provide an example of how metabolic exchange could lead to potential beneficial effects in the host. IMPORTANCE Microbial interactions represent the inner connections in the gut microbiome. By integrating mathematical modeling tools and microbial bidirectional culturing, we determined how two gut commensals engage in the exchange of cross-feeding metabolites, lactate and succinate, for increased growth in two fibers. These interactions underpinned butyrate production in cocultures, resulting in a significant reduction in cellular inflammation and protection against microbial toxins when applied to cellular models.
Collapse
|
109
|
Liu Y, Feng J, Pan H, Zhang X, Zhang Y. Genetically engineered bacterium: Principles, practices, and prospects. Front Microbiol 2022; 13:997587. [PMID: 36312915 PMCID: PMC9606703 DOI: 10.3389/fmicb.2022.997587] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/23/2022] [Indexed: 12/24/2022] Open
Abstract
Advances in synthetic biology and the clinical application of bacteriotherapy enable the use of genetically engineered bacteria (GEB) to combat various diseases. GEB act as a small 'machine factory' in the intestine or other tissues to continuously produce heterologous proteins or molecular compounds and, thus, diagnose or cure disease or work as an adjuvant reagent for disease treatment by regulating the immune system. Although the achievements of GEBs in the treatment or adjuvant therapy of diseases are promising, the practical implementation of this new therapeutic modality remains a grand challenge, especially at the initial stage. In this review, we introduce the development of GEBs and their advantages in disease management, summarize the latest research advances in microbial genetic techniques, and discuss their administration routes, performance indicators and the limitations of GEBs used as platforms for disease management. We also present several examples of GEB applications in the treatment of cancers and metabolic diseases and further highlight their great potential for clinical application in the near future.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Hangcheng Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xiuwei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Yunlei Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Department of Biomedical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
110
|
Markelova NN, Semenova EF, Sineva ON, Sadykova VS. The Role of Cyclomodulins and Some Microbial Metabolites in Bacterial Microecology and Macroorganism Carcinogenesis. Int J Mol Sci 2022; 23:ijms231911706. [PMID: 36233008 PMCID: PMC9570213 DOI: 10.3390/ijms231911706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
A number of bacteria that colonize the human body produce toxins and effectors that cause changes in the eukaryotic cell cycle—cyclomodulins and low-molecular-weight compounds such as butyrate, lactic acid, and secondary bile acids. Cyclomodulins and metabolites are necessary for bacteria as adaptation factors—which are influenced by direct selection—to the ecological niches of the host. In the process of establishing two-way communication with the macroorganism, these compounds cause limited damage to the host, despite their ability to disrupt key processes in eukaryotic cells, which can lead to pathological changes. Possible negative consequences of cyclomodulin and metabolite actions include their potential role in carcinogenesis, in particular, with the ability to cause DNA damage, increase genome instability, and interfere with cancer-associated regulatory pathways. In this review, we aim to examine cyclomodulins and bacterial metabolites as important factors in bacterial survival and interaction with the host organism to show their heterogeneous effect on oncogenesis depending on the surrounding microenvironment, pathological conditions, and host genetic background.
Collapse
Affiliation(s)
- Natalia N. Markelova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| | - Elena F. Semenova
- Institute of Biochemical Technology, Ecology and Pharmacy, V.I. Vernadsky Crimean Federal University, 295007 Simferopol, Russia
| | - Olga N. Sineva
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
| | - Vera S. Sadykova
- Gause Institute of New Antibiotics, ul. Bolshaya Pirogovskaya, 11, 119021 Moscow, Russia
- Correspondence: (N.N.M.); (V.S.S.)
| |
Collapse
|
111
|
Rekha K, Venkidasamy B, Samynathan R, Nagella P, Rebezov M, Khayrullin M, Ponomarev E, Bouyahya A, Sarkar T, Shariati MA, Thiruvengadam M, Simal-Gandara J. Short-chain fatty acid: An updated review on signaling, metabolism, and therapeutic effects. Crit Rev Food Sci Nutr 2022; 64:2461-2489. [PMID: 36154353 DOI: 10.1080/10408398.2022.2124231] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Fatty acids are good energy sources (9 kcal per gram) that aerobic tissues can use except for the brain (glucose is an alternative source). Apart from the energy source, fatty acids are necessary for cell signaling, learning-related memory, modulating gene expression, and functioning as cytokine precursors. Short-chain fatty acids (SCFAs) are saturated fatty acids arranged as a straight chain consisting minimum of 6 carbon atoms. SCFAs possess various beneficial effects like improving metabolic function, inhibiting insulin resistance, and ameliorating immune dysfunction. In this review, we discussed the biogenesis, absorption, and transport of SCFA. SCFAs can act as signaling molecules by stimulating G protein-coupled receptors (GPCRs) and suppressing histone deacetylases (HDACs). The role of SCFA on glucose metabolism, fatty acid metabolism, and its effect on the immune system is also reviewed with updated details. SCFA possess anticancer, anti-diabetic, and hepatoprotective effects. Additionally, the association of protective effects of SCFA against brain-related diseases, kidney diseases, cardiovascular damage, and inflammatory bowel diseases were also reviewed. Nanotherapy is a branch of nanotechnology that employs nanoparticles at the nanoscale level to treat various ailments with enhanced drug stability, solubility, and minimal side effects. The SCFA functions as drug carriers, and nanoparticles were also discussed. Still, much research was not focused on this area. SCFA functions in host gene expression through inhibition of HDAC inhibition. However, the study has to be focused on the molecular mechanism of SCFA against various diseases that still need to be investigated.
Collapse
Affiliation(s)
- Kaliaperumal Rekha
- Department of Environmental and Herbal Science, Tamil University, Thanjavur, Tamil Nadu, India
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | | | - Praveen Nagella
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, Karnataka, India
| | - Maksim Rebezov
- Department of Scientific Research, V. M. Gorbatov Federal Research Center for Food Systems, Moscow, Russia
- Department of Scientific Research, Russian State Agrarian University-Moscow Timiryazev Agricultural Academy, Moscow, Russia
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Mars Khayrullin
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Evgeny Ponomarev
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda, West Bengal, India
| | - Mohammad Ali Shariati
- Department of Scientific Research, Russian State Agrarian University-Moscow Timiryazev Agricultural Academy, Moscow, Russia
- Department of Scientific Research, K. G. Razumovsky Moscow State University of technologies and management (The First Cossack University), Moscow, Russia
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Sciences, Konkuk University, Seoul, South Korea
| | - Jesus Simal-Gandara
- Analytical Chemistry and Food Science Department, Faculty of Science, Universidade de Vigo, Nutrition and Bromatology Group, Ourense, Spain
| |
Collapse
|
112
|
Oleic Acid and Palmitic Acid from Bacteroides thetaiotaomicron and Lactobacillus johnsonii Exhibit Anti-Inflammatory and Antifungal Properties. Microorganisms 2022; 10:microorganisms10091803. [PMID: 36144406 PMCID: PMC9504516 DOI: 10.3390/microorganisms10091803] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/21/2022] Open
Abstract
A decrease in populations of Bacteroides thetaiotaomicron and Lactobacillus johnsonii is observed during the development of colitis and fungal overgrowth, while restoration of these populations reduces inflammatory parameters and fungal overgrowth in mice. This study investigated the effect of two fatty acids from B. thetaiotaomicron and L. johnsonii on macrophages and Caco-2 cells, as well as their impact on the inflammatory immune response and on Candida glabrata overgrowth in a murine model of dextran sulfate sodium (DSS)-induced colitis. Oleic acid (OA) and palmitic acid (PA) from L. johnsonii and B. thetaiotaomicron were detected during their interaction with epithelial cells from colon samples. OA alone or OA combined with PA (FAs) reduced the expression of proinflammatory mediators in intestinal epithelial Caco-2 cells challenged with DSS. OA alone or FAs increased FFAR1, FFAR2, AMPK, and IL-10 expression in macrophages. Additionally, OA alone or FAs decreased COX-2, TNFα, IL-6, and IL-12 expression in LPS-stimulated macrophages. In the DSS murine model, oral administration of FAs reduced inflammatory parameters, decreased Escherichia coli and Enterococcus faecalis populations, and eliminated C. glabrata from the gut. Overall, these findings provide evidence that OA combined with PA exhibits anti-inflammatory and antifungal properties.
Collapse
|
113
|
Detection of indigenous gut bacteria related to red chilli pepper (Capsicum annuum) in murine caecum and human faecal cultures. Mol Biol Rep 2022; 49:10239-10250. [PMID: 36068389 DOI: 10.1007/s11033-022-07875-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/15/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Red chilli pepper (Capsicum annuum; RP) is a popular spice containing the active compound capsaicin. Indigenous gut bacteria and metabolism can affect host health. The functions of capsaicin, including the regulation of metabolic health and anti-oxidant properties, may be correlated with the gut microbiota. METHODS To identify indigenous gut bacteria that are responsive to RP, Institute of Cancer Research mice fed a diet with no fibre or with 5% (w/w) RP for 14 days. Additionally, human stool samples collected from four healthy volunteers were incubated without (control) or with 2% (w/v) RP at 37 °C for 24 h. Microbiota in murine caecal samples and human faecal cultures were analysed using 16S rRNA (V4) amplicon sequencing. RESULTS Compared with the microbiota in mice fed no-fibre diets, Lachnospiraceae spp.-, Muribaculaceae spp.-, and Phacaeicola vulgatus-like bacteria were defined as murine RP-responsive indigenous gut bacteria (RP-RIB). In the human faecal cultures, acetate and propionate levels were higher in RP cultures than in the control cultures. Subdoligranulum spp.-, Blautia spp.-, Faecalibacterium prausnitzii-, P. vulgatus-, and Prevotella copri-like bacteria were defined as human RP-RIB. Compared with control culture Fe-reducing power was increased in the culture with RP. CONCLUSION RP increases the amount of short-chain fatty acid-producing bacteria and beneficial gut bacteria in mouse and human faecal cultures. Overall, RP could have a positive effect on the host by altering the gut microbiota.
Collapse
|
114
|
Peng K, Xia S, Xiao S, Yu Q. Short-chain fatty acids affect the development of inflammatory bowel disease through intestinal barrier, immunology, and microbiota: A promising therapy? J Gastroenterol Hepatol 2022; 37:1710-1718. [PMID: 35906780 DOI: 10.1111/jgh.15970] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 12/09/2022]
Abstract
Intestinal metabolites are attracting increasing interest, especially more and more studies have found they are closely related to diseases. Microbial fermentation of indigestible dietary fibers in the gut produces short chain fatty acids (SCFAs) as the main product. SCFAs can exert influences on the integrity of the intestinal epithelial and mucosal barrier, immune reactions, and the diversity of microbiota in humans. Thus, alteration in SCFAs may affect inflammatory bowel disease (IBD). In IBD, SCFAs are involved in the main pathogenic process and play an important role in the development of intestinal inflammation. Although many studies have proved that pretreatment with SCFAs can effectively ameliorate inflammation in the gut, the mechanisms are not fully understood. In this review, we describe the relationship between SCFAs and IBD from the aspects of defense barrier, immune effects, and microbial alterations. We also summarize the effects of SCFAs on comorbidities in IBD via the gut-brain, gut-liver, and gut-lung axis, and we give an overview of the prospects of their clinical application. A better understanding of the relevance of SCFAs in IBD may reveal novel targets for future study.
Collapse
Affiliation(s)
- Kaixin Peng
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suhong Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Xiao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Yu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
115
|
Li X, Peng B, Chi-Keung Cheung P, Wang J, Zheng X, You L. Depolymerized non-digestible sulfated algal polysaccharides produced by hydrothermal treatment with enhanced bacterial fermentation characteristics. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2022.107687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
116
|
Effects of Marine Bioactive Compounds on Gut Ecology Based on In Vitro Digestion and Colonic Fermentation Models. Nutrients 2022; 14:nu14163307. [PMID: 36014813 PMCID: PMC9412687 DOI: 10.3390/nu14163307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/30/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022] Open
Abstract
Digestion and the absorption of food compounds are necessary steps before nutrients can exert a role in human health. The absorption and utilization of nutrients in the diet is an extremely complex dynamic process. Accurately grasping the digestion and absorption mechanisms of different nutrients or bioactive compounds can provide a better understanding regarding the relationship between health and nutrition. Several in vitro models for simulating human gastrointestinal digestion and colonic fermentation have been established to obtain more accurate data for further understanding of the metabolism of dietary components. Marine media is rich in a wide variety of nutrients that are essential for humans and is gaining increased attention as a research topic. This review summarizes some of the most explored in vitro digestion and colonic fermentation models. It also summarizes the research progress on the digestion and absorption of nutrients and bioactive compounds from marine substrates when subjected to these in vitro models. Additionally, an overview of the changes imparted by the digestion process on these bioactive compounds is provided, in order to support those marine resources that can be utilized for developing new healthy foods.
Collapse
|
117
|
Moysidou CM, Withers AM, Nisbet AJ, Price DRG, Bryant CE, Cantacessi C, Owens RM. Investigation of Host-Microbe-Parasite Interactions in an In Vitro 3D Model of the Vertebrate Gut. Adv Biol (Weinh) 2022; 6:e2200015. [PMID: 35652159 DOI: 10.1002/adbi.202200015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/23/2022] [Indexed: 01/28/2023]
Abstract
In vitro models of the gut-microbiome axis are in high demand. Conventionally, intestinal monolayers grown on Transwell setups are used to test the effects of commensals/pathogens on the barrier integrity, both under homeostatic and pathophysiological conditions. While such models remain valuable for deepening the understanding of host-microbe interactions, often, they lack key biological components that mediate this intricate crosstalk. Here, a 3D in vitro model of the vertebrate intestinal epithelium, interfaced with immune cells surviving in culture for over 3 weeks, is developed and applied to proof-of-concept studies of host-microbe interactions. More specifically, the establishment of stable host-microbe cocultures is described and functional and morphological changes in the intestinal barrier induced by the presence of commensal bacteria are shown. Finally, evidence is provided that the 3D vertebrate gut models can be used as platforms to test host-microbe-parasite interactions. Exposure of gut-immune-bacteria cocultures to helminth "excretory/secretory products" induces in vivo-like up-/down-regulation of certain cytokines. These findings support the robustness of the modular in vitro cell systems for investigating the dynamics of host-microbe crosstalk and pave the way toward new approaches for systems biology studies of pathogens that cannot be maintained in vitro, including parasitic helminths.
Collapse
Affiliation(s)
- Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Aimee M Withers
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Alasdair J Nisbet
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Daniel R G Price
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Clare E Bryant
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| |
Collapse
|
118
|
Guo L, Guan Q, Duan W, Ren Y, Zhang XJ, Xu HY, Shi JS, Wang FZ, Lu R, Zhang HL, Xu ZH, Li H, Geng Y. Dietary Goji Shapes the Gut Microbiota to Prevent the Liver Injury Induced by Acute Alcohol Intake. Front Nutr 2022; 9:929776. [PMID: 35898713 PMCID: PMC9309278 DOI: 10.3389/fnut.2022.929776] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022] Open
Abstract
Diet is a major driver of the structure and function of the gut microbiota, which influences the host physiology. Alcohol abuse can induce liver disease and gut microbiota dysbiosis. Here, we aim to elucidate whether the well-known traditional health food Goji berry targets gut microbiota to prevent liver injury induced by acute alcohol intake. The results showed that Goji supplementation for 14 days alleviated acute liver injury as indicated by lowering serum aspartate aminotransferase, alanine aminotransferase, pro-inflammatory cytokines, as well as lipopolysaccharide content in the liver tissue. Goji maintained the integrity of the epithelial barrier and increased the levels of butyric acid in cecum contents. Furthermore, we established the causal relationship between gut microbiota and liver protection effects of Goji with the help of antibiotics treatment and fecal microbiota transplantation (FMT) experiments. Both Goji and FMT-Goji increased glutathione (GSH) in the liver and selectively enriched the butyric acid-producing gut bacterium Akkermansia and Ruminococcaceae by using 16S rRNA gene sequencing. Metabolomics analysis of cecum samples revealed that Goji and its trained microbiota could regulate retinoyl β-glucuronide, vanillic acid, and increase the level of glutamate and pyroglutamic acid, which are involved in GSH metabolism. Our study highlights the communication among Goji, gut microbiota, and liver homeostasis.
Collapse
Affiliation(s)
- Lin Guo
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Qijie Guan
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Wenhui Duan
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Yilin Ren
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiao-Juan Zhang
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Hong-Yu Xu
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Jin-Song Shi
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | | | - Ran Lu
- Ningxia Red Power Goji Co., Ltd, Zhongwei, China
| | - Hui-Ling Zhang
- Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety Control, Ningxia University, Yinchuan, China
| | - Zheng-Hong Xu
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
- Jiangsu Provincial Engineering Research Center for Bioactive Product Processing, Jiangnan University, Wuxi, China
| | - Huazhong Li
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
- *Correspondence: Huazhong Li
| | - Yan Geng
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
- Yan Geng
| |
Collapse
|
119
|
Xu Y, Milburn O, Beiersdorfer T, Du L, Akinbi H, Haslam DB. Antibiotic exposure prevents acquisition of beneficial metabolic functions in the preterm infant gut microbiome. MICROBIOME 2022; 10:103. [PMID: 35794664 PMCID: PMC9260971 DOI: 10.1186/s40168-022-01300-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Aberrations in the preterm microbiome following antibiotic therapy have been reported in previous studies. The objective of this study was to probe potential underlying mechanisms between this observation and susceptibility to adverse prematurity-related outcomes. RESULTS Metagenomic shotgun sequencing was performed on 133 stool and 253 skin samples collected at 1 and 3 weeks of age from 68 infants born at <36 weeks postmenstrual age and birth weight <2000 g. After accounting for gestational age and maternal antibiotics, the distribution of organisms in all samples and the corresponding metabolic pathway abundance were compared between infants exposed to postnatal antibiotics and antibiotics-naïve infants. In antibiotic-naïve infants, gestational and postnatal age imparted similar trajectories on maturation of the microbial community and associated metabolic functional capacity, with postnatal age exerting greater contribution. Antibiotic exposure was associated with reversal in maturation trajectory from the first week to the third week of age (p< 0.001). Butyrate-producing genera, including Clostridium and Blautia, were significantly more abundant in antibiotic-naïve neonates at 3 weeks postnatal age. Correspondingly, metabolic pathways required for short-chain fatty acid synthesis were significantly increased in antibiotic-naïve infants, but not in antibiotic-exposed neonates, at 3 weeks after birth. CONCLUSIONS Early brief antibiotic exposure markedly disrupts developmental trajectory of the neonatal microbiome and its corresponding functional capacity. Our findings may provide a mechanistic explanation for the known associations between antibiotic use and adverse outcomes in preterm infants. Video Abstract.
Collapse
Affiliation(s)
- Yanping Xu
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
- Global Health Center, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH USA
| | - Olivia Milburn
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
| | | | - Lizhong Du
- The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Henry Akinbi
- Perinatal Institute, CCHMC, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH USA
| | - David B. Haslam
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH USA
| |
Collapse
|
120
|
Wang X, Zhao J, Feng Y, Feng Z, Ye Y, Liu L, Kang G, Cao X. Evolutionary Insights Into Microbiota Transplantation in Inflammatory Bowel Disease. Front Cell Infect Microbiol 2022; 12:916543. [PMID: 35811664 PMCID: PMC9257068 DOI: 10.3389/fcimb.2022.916543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The intestinal microbiome plays an essential role in human health and disease status. So far, microbiota transplantation is considered a potential therapeutic approach for treating some chronic diseases, including inflammatory bowel disease (IBD). The diversity of gut microbiota is critical for maintaining resilience, and therefore, transplantation with numerous genetically diverse gut microbiota with metabolic flexibility and functional redundancy can effectively improve gut health than a single probiotic strain supplement. Studies have shown that natural fecal microbiota transplantation or washing microbiota transplantation can alleviate colitis and improve intestinal dysbiosis in IBD patients. However, unexpected adverse reactions caused by the complex and unclear composition of the flora limit its wider application. The evolving strain isolation technology and modifiable pre-existing strains are driving the development of microbiota transplantation. This review summarized the updating clinical and preclinical data of IBD treatments from fecal microbiota transplantation to washing microbiota transplantation, and then to artificial consortium transplantation. In addition, the factors considered for strain combination were reviewed. Furthermore, four types of artificial consortium transplant products were collected to analyze their combination and possible compatibility principles. The perspective on individualized microbiota transplantation was also discussed ultimately.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yuanhang Feng
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
| | - Zelin Feng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yulin Ye
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Limin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Guangbo Kang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- Institute of Shaoxing, Tianjin University, Zhejiang, China
- *Correspondence: Xiaocang Cao, ; Guangbo Kang,
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
- *Correspondence: Xiaocang Cao, ; Guangbo Kang,
| |
Collapse
|
121
|
Rashed R, Valcheva R, Dieleman LA. Manipulation of Gut Microbiota as a Key Target for Crohn's Disease. Front Med (Lausanne) 2022; 9:887044. [PMID: 35783604 PMCID: PMC9244564 DOI: 10.3389/fmed.2022.887044] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Crohn's disease (CD) is an inflammatory bowel disease (IBD) sub-type characterized by transmural chronic inflammation of the gastrointestinal tract. Research indicates a complex CD etiology involving genetic predisposition and immune dysregulation in response to environmental triggers. The chronic mucosal inflammation has been associated with a dysregulated state, or dysbiosis, of the gut microbiome (bacteria), mycobiome (fungi), virome (bacteriophages and viruses), and archeaome (archaea) further affecting the interkingdom syntrophic relationships and host metabolism. Microbiota dysbiosis in CD is largely described by an increase in facultative anaerobic pathobionts at the expense of strict anaerobic Firmicutes, such as Faecalibacterium prausnitzii. In the mycobiome, reduced fungal diversity and fungal-bacteria interactions, along with a significantly increased abundance of Candida spp. and a decrease in Saccharomyces cerevisiae are well documented. Virome analysis also indicates a significant decrease in phage diversity, but an overall increase in phages infecting bacterial groups associated with intestinal inflammation. Finally, an increase in methanogenic archaea such as Methanosphaera stadtmanae exhibits high immunogenic potential and is associated with CD etiology. Common anti-inflammatory medications used in CD management (amino-salicylates, immunomodulators, and biologics) could also directly or indirectly affect the gut microbiome in CD. Other medications often used concomitantly in IBD, such as antibiotics, antidepressants, oral contraceptives, opioids, and proton pump inhibitors, have shown to alter the gut microbiota and account for increased susceptibility to disease onset or worsening of disease progression. In contrast, some environmental modifications through alternative therapies including fecal microbiota transplant (FMT), diet and dietary supplements with prebiotics, probiotics, and synbiotics have shown potential protective effects by reversing microbiota dysbiosis or by directly promoting beneficial microbes, together with minimal long-term adverse effects. In this review, we discuss the different approaches to modulating the global consortium of bacteria, fungi, viruses, and archaea in patients with CD through therapies that include antibiotics, probiotics, prebiotics, synbiotics, personalized diets, and FMT. We hope to provide evidence to encourage clinicians and researchers to incorporate these therapies into CD treatment options, along with making them aware of the limitations of these therapies, and indicate where more research is needed.
Collapse
|
122
|
Xia Y, Kuda T, Nakamura S, Yamamoto M, Takahashi H, Kimura B. Effects of soy protein and β-conglycinin on microbiota and in vitro antioxidant and immunomodulatory capacities of human faecal cultures. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2022.107516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
123
|
Navarro-López V, Méndez-Miralles MÁ, Vela-Yebra R, Fríes-Ramos A, Sánchez-Pellicer P, Ruzafa-Costas B, Núñez-Delegido E, Gómez-Gómez H, Chumillas-Lidón S, Picó-Monllor JA, Navarro-Moratalla L. Gut Microbiota as a Potential Predictive Biomarker in Relapsing-Remitting Multiple Sclerosis. Genes (Basel) 2022; 13:genes13050930. [PMID: 35627315 PMCID: PMC9140870 DOI: 10.3390/genes13050930] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The influence of the microbiome on neurological diseases has been studied for years. Recent findings have shown a different composition of gut microbiota detected in patients with multiple sclerosis (MS). The role of this dysbiosis is still unknown. OBJECTIVE We analyzed the gut microbiota of 15 patients with active relapsing-remitting multiple sclerosis (RRMS), comparing with diet-matched healthy controls. METHOD To determine the composition of the gut microbiota, we performed high-throughput sequencing of the 16S ribosomal RNA gene. The specific amplified sequences were in the V3 and V4 regions of the 16S ribosomal RNA gene. RESULTS The gut microbiota of RRMS patients differed from healthy controls in the levels of the Lachnospiraceae, Ezakiella, Ruminococcaceae, Hungatella, Roseburia, Clostridium, Shuttleworthia, Poephyromonas, and Bilophila genera. All these genera were included in a logistic regression analysis to determine the sensitivity and the specificity of the test. Finally, the ROC (receiver operating characteristic) and AUC with a 95% CI were calculated and best-matched for Ezakiella (AUC of 75.0 and CI from 60.6 to 89.4) and Bilophila (AUC of 70.2 and CI from 50.1 to 90.4). CONCLUSIONS There is a dysbiosis in the gut microbiota of RRMS patients. An analysis of the components of the microbiota suggests the role of some genera as a predictive factor of RRMS prognosis and diagnosis.
Collapse
Affiliation(s)
- Vicente Navarro-López
- Ph.D. Program in Health Sciences, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
- Infectious Disease Unit, University Hospital Vinalopó, Carrer Tonico Sansano Mora 14, 03293 Elche, Spain
- Correspondence: (V.N.-L.); (M.Á.M.-M.)
| | - María Ángeles Méndez-Miralles
- Ph.D. Program in Health Sciences, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
- Department of Neurology, University Hospital of Torrevieja, Carretera CV95, s/n, 03186 Alicante, Spain;
- Correspondence: (V.N.-L.); (M.Á.M.-M.)
| | - Rosa Vela-Yebra
- Department of Neurology, University Hospital of Torrevieja, Carretera CV95, s/n, 03186 Alicante, Spain;
| | - Ana Fríes-Ramos
- Department of Neurology, University Hospital of Vinalopó, Carrer Tonico Sansano Mora 14, 03293 Elche, Spain;
| | - Pedro Sánchez-Pellicer
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
| | - Beatriz Ruzafa-Costas
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
| | - Eva Núñez-Delegido
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
| | - Humberto Gómez-Gómez
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
| | - Sara Chumillas-Lidón
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
| | - Jose A. Picó-Monllor
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
- Department of Pharmacology, Pediatrics and Organic Chemistry, Faculty of Pharmacy, Universidad Miguel Hernández de Elche, 03202 Elche, Spain
| | - Laura Navarro-Moratalla
- MiBioPath Research Group, Department of Clinical Medicine, Campus de los Jerónimos 135, UCAM-Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain; (P.S.-P.); (B.R.-C.); (E.N.-D.); (H.G.-G.); (S.C.-L.); (J.A.P.-M.); (L.N.-M.)
| |
Collapse
|
124
|
Abstract
Inflammatory bowel diseases (IBD), namely, Crohn's disease (CD) and ulcerative colitis (UC), are lifelong and incurable chronic inflammatory diseases affecting 6.8 million people worldwide. By 2030, the prevalence of IBD is estimated to reach 1% of the population in Western countries, and thus there is an urgent need to develop effective therapies to reduce the burden of this disease. Microbiome dysbiosis is at the heart of the IBD pathophysiology, and current research and development efforts for IBD treatments have been focused on gut microbiome regulation. Diet can shape the intestinal microbiome. Diet is also preferred over medication, is safe, and has been proven to be an effective strategy for the management of IBD. Therefore, although often overlooked, dietary interventions targeting the microbiome represent ideal treatments for IBD. Here, I summarize the latest research on diet as a treatment for IBD from infancy to adulthood, compile evidence of the mechanisms of action behind diet as treatment, and, lastly, provide insights into future research focusing on culturally tailored diets for ethnic minority groups with increased incidence of IBD yet underrepresented in nutrition research.
Collapse
Affiliation(s)
- Ana Maldonado-Contreras
- University of Massachusetts Chan Medical School, Department of Microbiology and Physiological Systems, Program of Microbiome Dynamics, Worcester, Massachusetts, USA
| |
Collapse
|
125
|
Ren X, Xing Y, He L, Xiu Z, Yang L, Han A, Jia Q, Dong Y. Effect of 1-Deoxynojirimycin on insulin resistance in prediabetic mice based on next-generation sequencing and intestinal microbiota study. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115029. [PMID: 35077826 DOI: 10.1016/j.jep.2022.115029] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 1-Deoxynojirimycin (DNJ), the major alkaloid in Morus alba L., is the main effective constituent in "Mulberry twig Alkaloids Tablets" launched in China in 2020. Prediabetes, characterized by insulin resistance, is regarded as the key period for reversing Type 2 diabetes mellitus (T2DM) through lifestyle intervention and glucose-lowering drugs. Besides the excellent activity as an α-glucosidase inhibitor, DNJ also improves insulin sensitivity in T2DM murine models, yet the mechanism is still unclear. Besides, the pharmaceutical effect of DNJ on prediabetes is also undocumented. AIM OF THE STUDY The aim of this study was to investigate the pharmaceutical effect of DNJ on high-fat and streptozotocin (STZ)-induced prediabetes mice, and to elucidate the mechanism of insulin resistance ameliorated by DNJ. MATERIALS AND METHODS Oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) were performed to detect blood glucose level and insulin sensitivity in mice. The levels of circulating lipopolysaccharide (LPS), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) in the plasma of mice were measured by limulus reagent and enzyme-linked immunosorbent assay (ELISA), respectively. Next-generation sequencing (NGS) and intestinal microbiota sequencing were used to screen the alterations in the transcriptome of liver tissues and to assess the differences in intestinal flora composition, respectively. Expression of cytokine signaling pathway inhibitor 3 (SOCS3), insulin receptor substrate (IRS1), p-IRS1 (Tyr896), occludin, and toll like receptor 4 (TLR4)/NF-κB signaling pathway were confirmed by western blotting. RESULTS Our study revealed that DNJ decreased the blood glucose level and improve insulin sensitivity in prediabetic mice. DNJ significantly reduced the relative risk of T2DM in prediabetic mice by approximately 83.7%. Mechanistically, DNJ treatment suppressed the circulating levels of LPS, IL-6, and TNF-α in plasma and decreased the inflammatory infiltration in liver and colon tissues. DNJ-treatment increased the abundance of Akkermansia, Bifidobacterium, and Lactobacillus, and decreased the abundance of Enterococcaceae and Lachnospiraceae. Moreover, DNJ suppressed the expression of SOCS3 and the activity of TLR4/NF-κB signaling pathway, meanwhile improving the expression of occludin and the ratio of p-IRS1 (Tyr896)/IRS1. CONCLUSIONS DNJ effectively ameliorates glucose and lipid metabolism in prediabetic mice, and decreased the relative risk of progression into T2DM from prediabetes. The suppressed immune responses play essential roles in the improvement of insulin resistance by DNJ treatment. In conclusion, DNJ from Morus alba L. is a promising alternative agent in T2DM prevention.
Collapse
Affiliation(s)
- Xinxiu Ren
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Yan Xing
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Liangyu He
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Zhilong Xiu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| | - Ling Yang
- Xinjiang Production and Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Tarim University, Alar, Xinjiang, 843300, China; Instrumental Analysis Center, Tarim University, Alar, Xinjiang, 843300, China.
| | - Aizhi Han
- Instrumental Analysis Center, Tarim University, Alar, Xinjiang, 843300, China.
| | - Qinhua Jia
- Instrumental Analysis Center, Tarim University, Alar, Xinjiang, 843300, China.
| | - Yuesheng Dong
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| |
Collapse
|
126
|
Olendzki B, Bucci V, Cawley C, Maserati R, McManus M, Olednzki E, Madziar C, Chiang D, Ward DV, Pellish R, Foley C, Bhattarai S, McCormick BA, Maldonado-Contreras A. Dietary manipulation of the gut microbiome in inflammatory bowel disease patients: Pilot study. Gut Microbes 2022; 14:2046244. [PMID: 35311458 PMCID: PMC8942410 DOI: 10.1080/19490976.2022.2046244] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Diet is a modifiable, noninvasive, inexpensive behavior that is crucial in shaping the intestinal microbiome. A microbiome "imbalance" or dysbiosis in inflammatory bowel disease (IBD) is linked to inflammation. Here, we aim to define the impact of specific foods on bacterial species commonly depleted in patients with IBD to better inform dietary treatment. We performed a single-arm, pre-post intervention trial. After a baseline period, a dietary intervention with the IBD-Anti-Inflammatory Diet (IBD-AID) was initiated. We collected stool and blood samples and assessed dietary intake throughout the study. We applied advanced computational approaches to define and model complex interactions between the foods reported and the microbiome. A dense dataset comprising 553 dietary records and 340 stool samples was obtained from 22 participants. Consumption of prebiotics, probiotics, and beneficial foods correlated with increased abundance of Clostridia and Bacteroides, commonly depleted in IBD cohorts. We further show that specific foods categorized as prebiotics or adverse foods are correlated to levels of cytokines in serum (i.e., GM-CSF, IL-6, IL-8, TNF-alpha) that play a central role in IBD pathogenesis. By using robust predictive analytics, this study represents the first steps to detangle diet-microbiome and diet-immune interactions to inform personalized nutrition for patients suffering from dysbiosis-related IBD.
Collapse
Affiliation(s)
- Barbara Olendzki
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Caitlin Cawley
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Rene Maserati
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Margaret McManus
- Center for Clinical and Translational Science, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Effie Olednzki
- Center for Applied Nutrition, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Camilla Madziar
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - David Chiang
- Department of Medicine,University of Massachusetts Medical SchoolWorcester, Massachusetts, USA
| | - Doyle V. Ward
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Randall Pellish
- UMass Memorial Medical Center University Campus, Department of Gastroenterology
| | - Christine Foley
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shakti Bhattarai
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ana Maldonado-Contreras
- Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics. University of Massachusetts Medical School, Worcester, Massachusetts, USA,CONTACT Ana Maldonado-Contreras Department of Microbiology and Physiological Systems and Program of Microbiome Dynamics, 368 Plantation Street, Albert Sherman Center, Office AS.81045, Worcester, Massachusetts, 01605, Worcester, Massachusetts, USA
| |
Collapse
|
127
|
Brochard C, Bouguen G, Olivier R, Durand T, Henno S, Peyronnet B, Pagenault M, Lefèvre C, Boudry G, Croyal M, Fautrel A, Esvan M, Ropert A, Dariel A, Siproudhis L, Neunlist M. Altered epithelial barrier functions in the colon of patients with spina bifida. Sci Rep 2022; 12:7196. [PMID: 35505001 PMCID: PMC9065040 DOI: 10.1038/s41598-022-11289-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/12/2022] [Indexed: 11/09/2022] Open
Abstract
Our objectives were to better characterize the colorectal function of patients with Spina Bifida (SB). Patients with SB and healthy volunteers (HVs) completed prospectively a standardized questionnaire, clinical evaluation, rectal barostat, colonoscopy with biopsies and faecal collection. The data from 36 adults with SB (age: 38.8 [34.1-47.2]) were compared with those of 16 HVs (age: 39.0 [31.0-46.5]). Compared to HVs, rectal compliance was lower in patients with SB (p = 0.01), whereas rectal tone was higher (p = 0.0015). Ex vivo paracellular permeability was increased in patients with SB (p = 0.0008) and inversely correlated with rectal compliance (r = - 0.563, p = 0.002). The expression of key tight junction proteins and inflammatory markers was comparable between SB and HVs, except for an increase in Claudin-1 immunoreactivity (p = 0.04) in SB compared to HVs. TGFβ1 and GDNF mRNAs were expressed at higher levels in patients with SB (p = 0.02 and p = 0.008). The levels of acetate, propionate and butyrate in faecal samples were reduced (p = 0.04, p = 0.01, and p = 0.02, respectively). Our findings provide evidence that anorectal and epithelial functions are altered in patients with SB. The alterations in these key functions might represent new therapeutic targets, in particular using microbiota-derived approaches.Clinical Trials: NCT02440984 and NCT03054415.
Collapse
Affiliation(s)
- Charlène Brochard
- Service d'Explorations Fonctionnelles Digestives, CHRU Pontchaillou, Université de Rennes 1, 2 rue Henri le Guillou, 35033, Rennes Cedex, France.
- The Enteric Nervous System in Gut and Brain Disorders INSERM, TENS, Université de Nantes, Nantes, France.
- Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), CHU Rennes, 35000, Rennes, France.
- Centre Référence Maladies Rares Spina Bifida, CHRU Pontchaillou, Rennes, France.
| | - Guillaume Bouguen
- Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), CHU Rennes, 35000, Rennes, France
- Service des Maladies de l'Appareil Digestif, CHRU Pontchaillou, Université de Rennes 1, Rennes, France
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France
| | - Raphael Olivier
- The Enteric Nervous System in Gut and Brain Disorders INSERM, TENS, Université de Nantes, Nantes, France
| | - Tony Durand
- The Enteric Nervous System in Gut and Brain Disorders INSERM, TENS, Université de Nantes, Nantes, France
| | - Sébastien Henno
- Service d'Anatomopathologie, CHRU Pontchaillou, Rennes, France
| | - Benoît Peyronnet
- Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), CHU Rennes, 35000, Rennes, France
- Centre Référence Maladies Rares Spina Bifida, CHRU Pontchaillou, Rennes, France
- Service d'Urologie, CHRU Pontchaillou, Rennes, France
| | - Mael Pagenault
- Service des Maladies de l'Appareil Digestif, CHRU Pontchaillou, Université de Rennes 1, Rennes, France
| | - Chloé Lefèvre
- The Enteric Nervous System in Gut and Brain Disorders INSERM, TENS, Université de Nantes, Nantes, France
| | - Gaëlle Boudry
- Institut Numecan, INSERM, INRAE, Univ Rennes, Rennes, France
| | - Mikael Croyal
- Université de Nantes, CHU Nantes, INSERM, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, 44000, Nantes, France
- CRNH-Ouest Mass Spectrometry Core Facility, 44000, Nantes, France
| | - Alain Fautrel
- Plateforme H2P2, Université de Rennes, Rennes, France
| | - Maxime Esvan
- Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), CHU Rennes, 35000, Rennes, France
| | - Alain Ropert
- Service d'Explorations Fonctionnelles Digestives, CHRU Pontchaillou, Université de Rennes 1, 2 rue Henri le Guillou, 35033, Rennes Cedex, France
| | - Anne Dariel
- Service de Chirurgie Pédiatrique, CHU Marseille, Marseille, France
| | - Laurent Siproudhis
- Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), CHU Rennes, 35000, Rennes, France
- Centre Référence Maladies Rares Spina Bifida, CHRU Pontchaillou, Rennes, France
- Service des Maladies de l'Appareil Digestif, CHRU Pontchaillou, Université de Rennes 1, Rennes, France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain Disorders INSERM, TENS, Université de Nantes, Nantes, France
| |
Collapse
|
128
|
Hu S, Mok J, Gowans M, Ong DEH, Hartono JL, Lee JWJ. Oral Microbiome of Crohn's Disease Patients With and Without Oral Manifestations. J Crohns Colitis 2022; 16:1628-1636. [PMID: 35511486 PMCID: PMC9624293 DOI: 10.1093/ecco-jcc/jjac063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/03/2022] [Accepted: 04/27/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Microbiome dysbiosis is associated with inflammatory destruction in Crohn's disease [CD]. Although gut microbiome dysbiosis is well established in CD, the oral microbiome is comparatively under-studied. This study aims to characterize the oral microbiome of CD patients with/without oral manifestations. METHODS Patients with CD were recruited with age-, gender- and race-matched controls. Potential confounders such as dental caries and periodontal condition were recorded. The oral microbiome was collected using saliva samples. Microbial DNA was extracted and sequenced using shotgun sequencing. Metagenomic taxonomic and functional profiles were generated and analysed. RESULTS The study recruited 41 patients with CD and 24 healthy controls. Within the CD subjects, 39.0% had oral manifestations with the majority presenting with cobblestoning and/or oral ulcers. Principal coordinate analysis demonstrated distinct oral microbiome profiles between subjects with and without CD, with four key variables responsible for overall oral microbiome variance: [1] diagnosis of CD, [2] concomitant use of steroids, [3] concomitant use of azathioprine and 4] presence of oral ulcers. Thirty-two significant differentially abundant microbial species were identified, with the majority associated with the diagnosis of CD. A predictive model based on differences in the oral microbiome found that the oral microbiome has strong discriminatory function to distinguish subjects with and without CD [AUROC 0.84]. Functional analysis found that an increased representation of microbial enzymes [n = 5] in the butyrate pathway was positively associated with the presence of oral ulcers. CONCLUSIONS The oral microbiome can aid in the diagnosis of CD and its composition was associated with oral manifestations.
Collapse
Affiliation(s)
- Shijia Hu
- Faculty of Dentistry, National University of Singapore, Singapore
| | - John Mok
- Division of Gastroenterology & Hepatology, National University Hospital, Singapore
| | - Michelle Gowans
- Division of Gastroenterology & Hepatology, National University Hospital, Singapore
| | - David E H Ong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Juanda Leo Hartono
- Division of Gastroenterology & Hepatology, National University Hospital, Singapore,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jonathan Wei Jie Lee
- Corresponding author: Jonathan Wei Jie Lee, Division of Gastroenterology & Hepatology, National University Hospital, Singapore, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore.
| |
Collapse
|
129
|
Xu T, Ma X, Zhou X, Qian M, Yang Z, Cao P, Han X. Coated tannin supplementation improves growth performance, nutrients digestibility, and intestinal function in weaned piglets. J Anim Sci 2022; 100:skac088. [PMID: 35298652 PMCID: PMC9109020 DOI: 10.1093/jas/skac088] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
To explore the effect of coated tannin (CT) on the growth performance, nutrients digestibility, and intestinal function in weaned piglets, a total of 180 piglets Duroc × Landrace × Yorkshire (28 d old) weighing about 8.6 kg were randomly allotted to three treatments: 1) Con: basal diet (contains ZnSO4); 2) Tan: basal diet + 0.15% CT; and 3) ZnO: basal diet + ZnO (Zn content is 1,600 mg/kg). The results showed that 0.15% CT could highly increase the average daily gain and average daily feed intake of weaned piglets compared with the control group, especially decreasing diarrhea incidence significantly (P < 0.05). Compared with the control group, crude protein apparent digestibility and digestive enzyme activity of the piglets fed with 0.15% CT were enhanced obviously (P < 0.05). Meanwhile, the intestinal villi and microvilli arranged more densely, while the content of serum diamine oxidase was decreased, and the protein expressions of zonula occludens-1 (ZO-1) and claudin-1 were significantly upregulated (P < 0.05). In addition, CT altered the structure of intestinal microbiota and augmented some butyrate-producing bacteria such as Ruminococcaceae and Megasphaera. PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States) analysis also showed that the abundances of pathways related to butyrate metabolism and tryptophan metabolism were increased; however, the function of lipopolysaccharide biosynthesis proteins was significantly decreased. The results demonstrated that 0.15% CT could improve growth performance, digestibility, and intestinal function of weaned piglets, and it had the potential to replace ZnO applied to farming.
Collapse
Affiliation(s)
- Tingting Xu
- The Key Laboratory of Animal Nutrition and Feed Science in East China of Ministry of Agriculture, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xin Ma
- The Key Laboratory of Animal Nutrition and Feed Science in East China of Ministry of Agriculture, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xinchen Zhou
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Zhejiang University, Sanya, Hainan 572025, China
| | - Mengqi Qian
- The Key Laboratory of Animal Nutrition and Feed Science in East China of Ministry of Agriculture, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zhiren Yang
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Zhejiang University, Sanya, Hainan 572025, China
| | - Peiwen Cao
- The Key Laboratory of Animal Nutrition and Feed Science in East China of Ministry of Agriculture, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xinyan Han
- The Key Laboratory of Animal Nutrition and Feed Science in East China of Ministry of Agriculture, College of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Hainan Institute of Zhejiang University, Yazhou Bay Science and Technology City, Zhejiang University, Sanya, Hainan 572025, China
| |
Collapse
|
130
|
Worby CJ, Schreiber HL, Straub TJ, van Dijk LR, Bronson RA, Olson BS, Pinkner JS, Obernuefemann CLP, Muñoz VL, Paharik AE, Azimzadeh PN, Walker BJ, Desjardins CA, Chou WC, Bergeron K, Chapman SB, Klim A, Manson AL, Hannan TJ, Hooton TM, Kau AL, Lai HH, Dodson KW, Hultgren SJ, Earl AM. Longitudinal multi-omics analyses link gut microbiome dysbiosis with recurrent urinary tract infections in women. Nat Microbiol 2022; 7:630-639. [PMID: 35505248 PMCID: PMC9136705 DOI: 10.1038/s41564-022-01107-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/18/2022] [Indexed: 12/13/2022]
Abstract
Recurrent urinary tract infections (rUTIs) are a major health burden worldwide, with history of infection being a significant risk factor. While the gut is a known reservoir for uropathogenic bacteria, the role of the microbiota in rUTI remains unclear. We conducted a year-long study of women with (n = 15) and without (n = 16) history of rUTI, from whom we collected urine, blood and monthly faecal samples for metagenomic and transcriptomic interrogation. During the study 24 UTIs were reported, with additional samples collected during and after infection. The gut microbiome of individuals with a history of rUTI was significantly depleted in microbial richness and butyrate-producing bacteria compared with controls, reminiscent of other inflammatory conditions. However, Escherichia coli gut and bladder populations were comparable between cohorts in both relative abundance and phylogroup. Transcriptional analysis of peripheral blood mononuclear cells revealed expression profiles indicative of differential systemic immunity between cohorts. Altogether, these results suggest that rUTI susceptibility is in part mediated through the gut-bladder axis, comprising gut dysbiosis and differential immune response to bacterial bladder colonization, manifesting in symptoms.
Collapse
Affiliation(s)
- Colin J Worby
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
| | - Henry L Schreiber
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Timothy J Straub
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
| | - Lucas R van Dijk
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
| | - Ryan A Bronson
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
| | - Benjamin S Olson
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Jerome S Pinkner
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chloe L P Obernuefemann
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vanessa L Muñoz
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexandra E Paharik
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Philippe N Azimzadeh
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Wen-Chi Chou
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
| | - Karla Bergeron
- Department of Surgery, Division of Urologic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Sinéad B Chapman
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
| | - Aleksandra Klim
- Department of Surgery, Division of Urologic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Abigail L Manson
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA
| | - Thomas J Hannan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Andrew L Kau
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Division of Allergy and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - H Henry Lai
- Department of Surgery, Division of Urologic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen W Dodson
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Scott J Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA.
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
131
|
Lee G, Harada M, Midorikawa Y, Yamamoto M, Nakamura A, Takahashi H, Kuda T. Effects of alginate and laminaran on the microbiome and antioxidant properties of human faecal cultures. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
132
|
Garabatos N, Santamaria P. Gut Microbial Antigenic Mimicry in Autoimmunity. Front Immunol 2022; 13:873607. [PMID: 35572569 PMCID: PMC9094498 DOI: 10.3389/fimmu.2022.873607] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota plays a major role in the developmental biology and homeostasis of cells belonging to the adaptive and innate arms of the immune system. Alterations in its composition, which are known to be regulated by both genetic and environmental factors, can either promote or suppress the pathogenic processes underlying the development of various autoimmune diseases, including inflammatory bowel disease, multiple sclerosis, systemic lupus erythematosus, type 1 diabetes and rheumatoid arthritis, to just name a few. Cross-recognition of gut microbial antigens by autoreactive T cells as well as gut microbe-driven alterations in the activation and homeostasis of effector and regulatory T cells have been implicated in this process. Here, we summarize our current understanding of the positive and negative associations between alterations in the composition of the gut microbiota and the development of various autoimmune disorders, with a special emphasis on antigenic mimicry.
Collapse
Affiliation(s)
- Nahir Garabatos
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pere Santamaria
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Julia McFarlane Diabetes Research Centre (JMDRC), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
133
|
He P, Yu L, Tian F, Zhang H, Chen W, Zhai Q. Dietary Patterns and Gut Microbiota: The Crucial Actors in Inflammatory Bowel Disease. Adv Nutr 2022; 13:1628-1651. [PMID: 35348593 PMCID: PMC9526834 DOI: 10.1093/advances/nmac029] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
It is widely believed that diet and the gut microbiota are strongly related to the occurrence and progression of inflammatory bowel disease (IBD), but the effects of the interaction between dietary patterns and the gut microbiota on IBD have not been well elucidated. In this article, we aim to explore the complex relation between dietary patterns, gut microbiota, and IBD. We first comprehensively summarized the dietary patterns associated with IBD and found that dietary patterns can modulate the occurrence and progression of IBD through various signaling pathways, including mammalian target of rapamycin (mTOR), mitogen-activated protein kinases (MAPKs), signal transducer and activator of transcription 3 (STAT3), and NF-κB. Besides, the gut microbiota performs a vital role in the progression of IBD, which can affect the expression of IBD susceptibility genes, such as dual oxidase 2 (DUOX2) and APOA-1 , the intestinal barrier (in particular, the expression of tight junction proteins), immune function (especially the homeostasis between effector and regulatory T cells) and the physiological metabolism, in particular, SCFAs, bile acids (BAs), and tryptophan metabolism. Finally, we reviewed the current knowledge on the interaction between dietary patterns and the gut microbiota in IBD and found that dietary patterns modulate the onset and progression of IBD, which is partly attributed to the regulation of the gut microbiota (especially SCFAs-producing bacteria and Escherichia coli). Faecalibacteria as "microbiomarkers" of IBD could be used as a target for dietary interventions to alleviate IBD. A comprehensive understanding of the interplay between dietary intake, gut microbiota, and IBD will facilitate the development of personalized dietary strategies based on the regulation of the gut microbiota in IBD and expedite the era of precision nutritional interventions for IBD.
Collapse
Affiliation(s)
- Pandi He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China,Wuxi Translational Medicine Research Center, Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | | |
Collapse
|
134
|
Ruscogenins Improve CD-Like Enteritis by Inhibiting Apoptosis of Intestinal Epithelial Cells and Activating Nrf2/NQO1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4877275. [PMID: 35308175 PMCID: PMC8930266 DOI: 10.1155/2022/4877275] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 01/22/2022] [Accepted: 01/30/2022] [Indexed: 12/14/2022]
Abstract
Interaction of intestinal barrier dysfunction and intestinal inflammation promotes the progression of Crohn's disease (CD). A more recent study has suggested that ruscogenins (RUS) can exert anti-inflammatory effects through activation of the Nrf2/NQO1 pathway. The current study is aimed at determining the functionalization of RUS on CD-like colitis. Wild-type (WT) mice induced with trinitrobenzene sulfonic acid (TNBS) exhibit a significant inflammation in their colon and are hence widely used for CD models. In the current study, the mice were treated with the Nrf-2 antagonist (ML385) or ruscogenin (RUS) whereas normal WT mice were kept as the negative control. Comparative analysis was then performed on the inflammation and barrier function of the colons. In vitro analysis of mouse colonic organoid systems revealed the influence of RUS on LPS-induced apoptosis, cytokine, and chemokine expressions in the intestinal epithelium. It was found that RUS ameliorates murine colitis through activation of the Nrf2/NQO1 pathway which was presented as a decrease in inflammation score and downregulated levels of cytokine and chemokine synthesis, as well as increased intestinal permeability. Further, it was noted that RUS alleviated LPS-induced apoptosis in the intestinal epithelium cells through upregulation of the Nrf2/NQO1 signaling pathway in the mouse colonic organoids. In addition, ruscogenin (RUS) attenuated the levels of Bax and C-caspase-3 through activation of the Nrf2/HO1 signaling pathway both in vivo and in vitro. Therefore, it was evident that RUS can be applied as a potential alternative therapeutic agent in CD based on its protective effects on the barrier function and anti-inflammatory activity.
Collapse
|
135
|
Mukhopadhyay S, Saha S, Chakraborty S, Prasad P, Ghosh A, Aich P. Differential colitis susceptibility of Th1- and Th2-biased mice: A multi-omics approach. PLoS One 2022; 17:e0264400. [PMID: 35263357 PMCID: PMC8906622 DOI: 10.1371/journal.pone.0264400] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/09/2022] [Indexed: 01/08/2023] Open
Abstract
The health and economic burden of colitis is increasing globally. Understanding the role of host genetics and metagenomics is essential to establish the molecular basis of colitis pathogenesis. In the present study, we have used a common composite dose of DSS to compare the differential disease severity response in C57BL/6 (Th1 biased) and BALB/c (Th2 biased) mice with zero mortality rates. We employed multi-omics approaches and developed a newer vector analysis approach to understand the molecular basis of the disease pathogenesis. In the current report, comparative transcriptomics, metabonomics, and metagenomics analyses revealed that the Th1 background of C57BL/6 induced intense inflammatory responses throughout the treatment period. On the contrary, the Th2 background of BALB/c resisted severe inflammatory responses by modulating the host’s inflammatory, metabolic, and gut microbial profile. The multi-omics approach also helped us discover some unique metabolic and microbial markers associated with the disease severity. These biomarkers could be used in diagnostics.
Collapse
Affiliation(s)
- Sohini Mukhopadhyay
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - Subha Saha
- Institute of Life Sciences, NALCO Square, Bhubaneswar, Odisha, India
| | - Subhayan Chakraborty
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- School of Chemical Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| | - Punit Prasad
- Institute of Life Sciences, NALCO Square, Bhubaneswar, Odisha, India
| | - Arindam Ghosh
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- School of Chemical Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| |
Collapse
|
136
|
Giambò F, Costa C, Teodoro M, Fenga C. Role-Playing Between Environmental Pollutants and Human Gut Microbiota: A Complex Bidirectional Interaction. Front Med (Lausanne) 2022; 9:810397. [PMID: 35252248 PMCID: PMC8888443 DOI: 10.3389/fmed.2022.810397] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
There is a growing interest in the characterization of the involvement of toxicant and pollutant exposures in the development and the progression of several diseases such as obesity, diabetes, cancer, as well as in the disruption of the immune and reproductive homeostasis. The gut microbiota is considered a pivotal player against the toxic properties of chemicals with the establishment of a dynamic bidirectional relationship, underlining the toxicological significance of this mutual interplay. In fact, several environmental chemicals have been demonstrated to affect the composition, the biodiversity of the intestinal microbiota together with the underlining modulated metabolic pathways, which may play an important role in tailoring the microbiotype of an individual. In this review, we aimed to discuss the latest updates concerning the environmental chemicals–microbiota dual interaction, toward the identification of a distinctiveness of the gut microbial community, which, in turn, may allow to adopt personalized preventive strategies to improve risk assessment for more susceptible workers.
Collapse
Affiliation(s)
- Federica Giambò
- Occupational Medicine Section, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Chiara Costa
- Clinical and Experimental Medicine Department, University of Messina, Messina, Italy
| | - Michele Teodoro
- Occupational Medicine Section, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Concettina Fenga
- Occupational Medicine Section, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
137
|
Analysis of Gut Microbiota in Patients with Exacerbated Symptoms of Schizophrenia following Therapy with Amisulpride: A Pilot Study. Behav Neurol 2022; 2022:4262094. [PMID: 35287288 PMCID: PMC8917950 DOI: 10.1155/2022/4262094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/18/2022] [Indexed: 11/24/2022] Open
Abstract
Evidence is mounting that the gut microbiome is related to the underlying pathogenesis of schizophrenia. However, effects of amisulpride on gut microbiota are poorly defined. This study was aimed at analyzing cytokines and fecal microbiota in patients with exacerbated symptoms of schizophrenia treated with amisulpride during four weeks of their hospital stay. In the present study, feces collected from patients with schizophrenia were analyzed using 16S rRNA pyrosequencing and bioinformatic analyses to ascertain gut microbiome composition and fasting peripheral blood cytokines. We found that patients undergoing treatment of schizophrenia with amisulpride had distinct changes in gut microbial composition at the genus level, increased levels of short-chain fatty acid-producing bacteria (Dorea and Butyricicoccus), and reduced levels of pathogenic bacteria (Actinomyces and Porphyromonas), but the level of Desulfovibrio was still high. We also found a significant downregulation of butanoate metabolism based on functional analysis of the microbiome. After treatment, elevated levels of interleukin- (IL-) 4 and decreased levels of IL-6 were found. Our findings extend prior work and suggest a possible pharmacological mechanism of amisulpride treatment for schizophrenia, which acts via mediation of the gut microbiome.
Collapse
|
138
|
Loman BR, Russart KLG, Grant CV, Lynch AJ, Bailey MT, Pyter LM. Mammary tumors alter the fecal bacteriome and permit enteric bacterial translocation. BMC Cancer 2022; 22:245. [PMID: 35248004 PMCID: PMC8897840 DOI: 10.1186/s12885-022-09274-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 02/09/2022] [Indexed: 11/10/2022] Open
Abstract
Background Cancer patients experience gastrointestinal and behavioral symptoms, and are at increased risk of systemic infection and inflammation. These conditions are a major source of morbidity and decreased quality of life prior to cancer treatment, but poorly defined etiologies impede successful treatment. The gastrointestinal microbiota shape inflammation, influence cancer progression and treatment, and colonize tumors. However, research has not directly determined if peripheral tumors influence the microbiome and intestinal physiology, thus influencing gastrointestinal and behavioral symptoms. Therefore, the purpose of this study was to examine consequences of orthotopic, syngeneic mammary tumor implantation, growth, and resection on fecal bacteriome composition and intestinal barrier function in relation to systemic inflammation and enteric bacterial translocation in mice. Methods Female mice were randomized to 3 experimental groups: sham surgical control, tumor recipients, and tumor recipients later receiving tumor-resection. Mice were sacrificed three weeks after tumor implantation or resection for collection of stool, colon, spleen, and brain tissue and analysis. Results Tumor-bearing mice exhibited several markers of colonic barrier disruption, including dampened expression of tight junction proteins (Cldn1 and Ocln) and elevated circulating lipopolysaccharide binding protein (LBP). Compromised colonic barrier integrity was associated with altered fecal bacterial profiles in tumor-mice, including lower relative abundance of Lactobacillus, but higher Bacteroides. Consistent with colonic barrier disruption and altered microbiomes, tumor-mice displayed markers of systemic inflammation including splenomegaly, higher splenic bacterial load, and elevated splenic and brain pro-inflammatory cytokines. Several bacteria cultured from spleens had 16S rRNA gene amplicons matching those in fecal samples, suggesting they were of intestinal origin. Fecal Lactobacillus was highly-interrelated to physiological parameters disrupted by tumors via correlation network analysis. Tumor resection ameliorated circulating LBP, splenomegaly, and splenic cytokines, but not other parameters associated with loss of colonic barrier integrity and bacterial translocation. Conclusions Orthotopic mammary tumors alter the microbiome, reduce intestinal barrier function, increase translocation of enteric bacteria, and alter systemic inflammation. This provides insight into how tumors commence gastrointestinal and behavioral symptoms prior to treatment, and identify targets for future therapeutics, such as probiotic Lactobacillus supplementation. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09274-0.
Collapse
|
139
|
Doenyas C. Potential Role of Epigenetics and Redox Signaling in the Gut-Brain Communication and the Case of Autism Spectrum Disorder. Cell Mol Neurobiol 2022; 42:483-487. [PMID: 34773541 PMCID: PMC11441204 DOI: 10.1007/s10571-021-01167-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022]
Abstract
The gut-brain axis refers to the bidirectional connection and communication between the gastrointestinal tract and the central nervous system. This paper explores two routes for this communication that have hitherto remained under-examined: epigenetics and redox signaling and their implications for autism spectrum disorder (ASD). The gut microbiota may induce epigenetic changes in the gut and potentially in the brain through their fermentation products. Instead of through other conceptualizations of them acting as neurotransmitters, gut microbial products may act as epigenetic agents, which are supported by the effects of gut bacterial-derived metabolites on gene regulation and expression. In addition to their epigenetic effects, gut bacterial-derived communicative agents can also influence host signaling by contributing to and even substituting host reactive oxygen species (ROS) production. These ROS can act as second messengers and exert oxidative activity on proteins to influence immune, inflammatory, and other signaling processes. ROS and epigenetic mechanisms may have interactive effects as well. ROS, in addition to their role in signaling pathways and cellular redox alterations, also influence redox-sensitive transcription factors, thereby having an effect on gene expression. Specifically, ROS are involved in the activation of transcription factors, chromatin remodeling, and histone/protein deacetylation. These two proposed mechanisms correspond with the recent findings related to ASD, where a cofactor that is shown to be lower in ASD has antioxidative properties, responds to epigenetic modulation, and increases via microbiota interventions. The current evidence reviewed here suggests the need to update models of the gut-brain communication to include these two mechanisms. Such a modeling can also contribute to understanding the unknowns of host metabolism and physiology in ASD and afford potential therapeutic avenues for this as well as other psychiatric and physiological conditions.
Collapse
|
140
|
Zhang S, Chen Q, Kelly CR, Kassam Z, Qin H, Li N, Tian H, Yang B, Zhao D, Ye C, Lin Z, Cui J, Zhou S, Chen X, Lv X, Yang R. Donor Screening for Fecal Microbiota Transplantation in China: Evaluation of 8483 Candidates. Gastroenterology 2022; 162:966-968.e3. [PMID: 34752816 DOI: 10.1053/j.gastro.2021.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/24/2021] [Accepted: 11/02/2021] [Indexed: 12/02/2022]
Affiliation(s)
- Shaoyi Zhang
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Qiyi Chen
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China.
| | - Colleen R Kelly
- Division of Gastroenterology, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Zain Kassam
- Finch Therapeutics, Somerville, Massachusetts
| | - Huanlong Qin
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China.
| | - Ning Li
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China
| | -
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hongliang Tian
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bo Yang
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Di Zhao
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chen Ye
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhiliang Lin
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jiaqu Cui
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shailan Zhou
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xia Chen
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xiaoqiong Lv
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rong Yang
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Tenth People's Hospital of Tongji University, Shanghai, China; Jianyi Yin, Department of Internal Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
141
|
Kaźmierczak-Siedlecka K, Skonieczna-Żydecka K, Hupp T, Duchnowska R, Marek-Trzonkowska N, Połom K. Next-generation probiotics - do they open new therapeutic strategies for cancer patients? Gut Microbes 2022; 14:2035659. [PMID: 35167406 PMCID: PMC8855854 DOI: 10.1080/19490976.2022.2035659] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gut microbiota and its association with cancer development/treatment has been intensively studied during the past several years. Currently, there is a growing interest toward next-generation probiotics (NGPs) as therapeutic agents that alter gut microbiota and impact on cancer development. In the present review we focus on three emerging NGPs, namely Faecalibacterium prausnitzii, Akkermansia muciniphila, and Bacteroides fragilis as their presence in the digestive tract can have an impact on cancer incidence. These NGPs enhance gastrointestinal immunity, maintain intestinal barrier integrity, produce beneficial metabolites, act against pathogens, improve immunotherapy efficacy, and reduce complications associated with chemotherapy and radiotherapy. Notably, the use of NGPs in cancer patients does not have a long history and, although their safety remains relatively undefined, recently published data has shown that they are non-toxigenic. Notwithstanding, A. muciniphila may promote colitis whereas enterotoxigenic B. fragilis stimulates chronic inflammation and participates in colorectal carcinogenesis. Nevertheless, the majority of B. fragilis strains provide a beneficial effect to the host, are non-toxigenic and considered as the best current NGP candidate. Overall, emerging studies indicate a beneficial role of these NGPs in the prevention of carcinogenesis and open new promising therapeutic options for cancer patients.
Collapse
Affiliation(s)
- Karolina Kaźmierczak-Siedlecka
- Department of Surgical Oncology, Medical University of Gdansk, Gdańsk, Poland,CONTACT Karolina Kaźmierczak-Siedlecka Department of Surgical Oncology, Medical University of Gdansk, Ul. Smoluchowskiego 18, 80-214Gdańsk, Poland
| | | | - Theodore Hupp
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland,Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | - Renata Duchnowska
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science University of Gdańsk, Gdańsk, Poland,Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Karol Połom
- Department of Surgical Oncology, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
142
|
Pre-Administration of Berberine Exerts Chemopreventive Effects in AOM/DSS-Induced Colitis-Associated Carcinogenesis Mice via Modulating Inflammation and Intestinal Microbiota. Nutrients 2022; 14:nu14040726. [PMID: 35215376 PMCID: PMC8879943 DOI: 10.3390/nu14040726] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
Inflammatory activation and intestinal flora imbalance play an essential role in the development and progression of colorectal cancer (CRC). Berberine (BBR) has attracted great attention in recent years due to its heath-related benefits in inflammatory disorders and tumors, but the intricate mechanisms have not been fully elucidated. In this study, the effects and the mechanism of BBR on colon cancer were investigated in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colitis-associated carcinogenesis mice model. Our results showed that pre-administration of BBR showed a decrease in weight loss, disease activity index (DAI) score, and the number of colon tumors in mice, compared with the model group. The evidence from pathological examination indicated that the malignancy of intestinal tumors was ameliorated after pre-administration of BBR. Additionally, pre-administration with BBR suppressed the expression of pro-inflammatory factors (interleukin (IL)-6, IL-1β, cyclooxygenase (COX)-2 and tumor necrosis factor (TNF)-α) and the cell-proliferation marker Ki67, while expression of the tight junction proteins (ZO-1 and occludin) were increased in colon tissue. Moreover, the levels of critical pathway proteins involved in the inflammatory process (p-STAT3 and p-JNK) and cell cycle regulation molecules (β-catenin, c-Myc and CylinD1) exhibited lower expression levels. Besides, 16S rRNA sequence analysis indicated that pre-administration of BBR increased the ratio of Firmicutes/Bacteroidetes (F:M) and the relative abundance of potentially beneficial bacteria, while the abundance of cancer-related bacteria was decreased. Gavage with Lactobacillus rhamnosus can improve the anti-tumor effect of BBR. Overall, pre-administration of BBR exerts preventive effects in colon carcinogenesis, and the mechanisms underlying these effects are correlated with the inhibition of inflammation and tumor proliferation and the maintenance of intestinal homeostasis.
Collapse
|
143
|
DeMartino P, Johnston EA, Petersen KS, Kris-Etherton PM, Cockburn DW. Additional Resistant Starch from One Potato Side Dish per Day Alters the Gut Microbiota but Not Fecal Short-Chain Fatty Acid Concentrations. Nutrients 2022; 14:nu14030721. [PMID: 35277080 PMCID: PMC8840755 DOI: 10.3390/nu14030721] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 01/11/2023] Open
Abstract
The composition of the gut microbiota and their metabolites are associated with cardiometabolic health and disease risk. Intake of dietary fibers, including resistant starch (RS), has been shown to favorably affect the health of the gut microbiome. The aim of this research was to measure changes in the gut microbiota and fecal short-chain fatty acids as part of a randomized, crossover supplemental feeding study. Fifty participants (68% female, aged 40 ± 13 years, BMI 24.5 ± 3.6 kg/m2) completed this study. Potato dishes (POT) contained more RS than refined grain dishes (REF) (POT: 1.31% wet basis (95% CI: 0.94, 1.71); REF: 0.73% wet basis (95% CI: 0.34, 1.14); p = 0.03). Overall, potato dish consumption decreased alpha diversity, but beta diversity was not impacted. Potato dish consumption was found to increase the abundance of Hungatella xylanolytica, as well as that of the butyrate producing Roseburia faecis, though fecal butyrate levels were unchanged. Intake of one potato-based side dish per day resulted in modest changes in gut microbiota composition and diversity, compared to isocaloric intake of refined grains in healthy adults. Studies examining foods naturally higher in RS are needed to understand microbiota changes in response to dietary intake of RS and associated health effects.
Collapse
Affiliation(s)
- Peter DeMartino
- Department of Food Science, Pennsylvania State University, University Park, PA 16802, USA;
| | - Emily A. Johnston
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA; (E.A.J.); (K.S.P.); (P.M.K.-E.)
| | - Kristina S. Petersen
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA; (E.A.J.); (K.S.P.); (P.M.K.-E.)
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Penny M. Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA 16802, USA; (E.A.J.); (K.S.P.); (P.M.K.-E.)
| | - Darrell W. Cockburn
- Department of Food Science, Pennsylvania State University, University Park, PA 16802, USA;
- Correspondence: ; Tel.: +1-814-863-2950
| |
Collapse
|
144
|
Zhang F, Wan Y, Zuo T, Yeoh YK, Liu Q, Zhang L, Zhan H, Lu W, Xu W, Lui GC, Li AY, Cheung CP, Wong CK, Chan PK, Chan FK, Ng SC. Prolonged Impairment of Short-Chain Fatty Acid and L-Isoleucine Biosynthesis in Gut Microbiome in Patients With COVID-19. Gastroenterology 2022; 162:548-561.e4. [PMID: 34687739 PMCID: PMC8529231 DOI: 10.1053/j.gastro.2021.10.013] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/16/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with altered gut microbiota composition. Phylogenetic groups of gut bacteria involved in the metabolism of short chain fatty acids (SCFAs) were depleted in SARS-CoV-2-infected patients. We aimed to characterize a functional profile of the gut microbiome in patients with COVID-19 before and after disease resolution. METHODS We performed shotgun metagenomic sequencing on fecal samples from 66 antibiotics-naïve patients with COVID-19 and 70 non-COVID-19 controls. Serial fecal samples were collected (at up to 6 times points) during hospitalization and beyond 1 month after discharge. We assessed gut microbial pathways in association with disease severity and blood inflammatory markers. We also determined changes of microbial functions in fecal samples before and after disease resolution and validated these functions using targeted analysis of fecal metabolites. RESULTS Compared with non-COVID-19 controls, patients with COVID-19 with severe/critical illness showed significant alterations in gut microbiome functionality (P < .001), characterized by impaired capacity of gut microbiome for SCFA and L-isoleucine biosynthesis and enhanced capacity for urea production. Impaired SCFA and L-isoleucine biosynthesis in gut microbiome persisted beyond 30 days after recovery in patients with COVID-19. Targeted analysis of fecal metabolites showed significantly lower fecal concentrations of SCFAs and L-isoleucine in patients with COVID-19 before and after disease resolution. Lack of SCFA and L-isoleucine biosynthesis significantly correlated with disease severity and increased plasma concentrations of CXCL-10, NT- proB-type natriuretic peptide, and C-reactive protein (all P < .05). CONCLUSIONS Gut microbiome of patients with COVID-19 displayed impaired capacity for SCFA and L-isoleucine biosynthesis that persisted even after disease resolution. These 2 microbial functions correlated with host immune response underscoring the importance of gut microbial functions in SARS-CoV-2 infection pathogenesis and outcome.
Collapse
Affiliation(s)
- Fen Zhang
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yating Wan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Tao Zuo
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yun Kit Yeoh
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Qin Liu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Lin Zhang
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hui Zhan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wenqi Lu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wenye Xu
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Grace C.Y. Lui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Amy Y.L. Li
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chun Pan Cheung
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chun Kwok Wong
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Paul K.S. Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Microbiology, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Francis K.L. Chan
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Microbiota I-Center (MagIC), Shatin, Hong Kong, China
| | - Siew C. Ng
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China,Microbiota I-Center (MagIC), Shatin, Hong Kong, China,Correspondence Address correspondence to: Siew C. Ng, PhD, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, 9/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Shatin, Hong Kong
| |
Collapse
|
145
|
Silva-Andrade C, Martin AJ, Garrido D. Comparative Genomics of Clostridium baratii Reveals Strain-Level Diversity in Toxin Abundance. Microorganisms 2022; 10:microorganisms10020213. [PMID: 35208668 PMCID: PMC8879937 DOI: 10.3390/microorganisms10020213] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/29/2021] [Accepted: 11/10/2021] [Indexed: 01/27/2023] Open
Abstract
Clostridium baratii strains are rare opportunistic pathogens associated with botulism intoxication. They have been isolated from foods, soil and be carried asymptomatically or cause botulism outbreaks. Is not taxonomically related to Clostridium botulinum, but some strains are equipped with BoNT/F7 cluster. Despite their relationship with diseases, our knowledge regarding the genomic features and phylogenetic characteristics is limited. We analyzed the pangenome of C. baratii to understand the diversity and genomic features of this species. We compared existing genomes in public databases, metagenomes, and one newly sequenced strain isolated from an asymptomatic subject. The pangenome was open, indicating it comprises genetically diverse organisms. The core genome contained 28.49% of the total genes of the pangenome. Profiling virulence factors confirmed the presence of phospholipase C in some strains, a toxin capable of disrupting eukaryotic cell membranes. Furthermore, the genomic analysis indicated significant horizontal gene transfer (HGT) events as defined by the presence of prophage genomes. Seven strains were equipped with BoNT/F7 cluster. The active site was conserved in all strains, identifying a missing 7-aa region upstream of the active site in C. baratii genomes. This analysis could be important to advance our knowledge regarding opportunistic clostridia and better understand their contribution to disease.
Collapse
Affiliation(s)
- Claudia Silva-Andrade
- Laboratorio de Biología de Redes, Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago 8580000, Chile;
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Alberto J. Martin
- Laboratorio de Biología de Redes, Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago 8580000, Chile;
- Correspondence: (A.J.M.); (D.G.)
| | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Correspondence: (A.J.M.); (D.G.)
| |
Collapse
|
146
|
El-Hakim Y, Bake S, Mani KK, Sohrabji F. Impact of intestinal disorders on central and peripheral nervous system diseases. Neurobiol Dis 2022; 165:105627. [PMID: 35032636 DOI: 10.1016/j.nbd.2022.105627] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/03/2022] [Accepted: 01/09/2022] [Indexed: 12/16/2022] Open
Abstract
Brain injuries and neurological diseases have a significant impact on the gut microbiome and the gut barrier. Reciprocally, gut disorders, such as Inflammatory Bowel Syndromes (IBS), can affect the development and pathology of neurodegenerative and neuropsychiatric diseases, although this aspect is less well studied and is the focus of this review. Inflammatory Bowel Syndrome (IBS) is a chronic and debilitating functional gastrointestinal disorder afflicting an estimated 9-23% of the world's population. A hallmark of this disease is leaky gut, a pathology in which the integrity of the gut blood barrier is compromised, causing gut contents such as immune cells and microbiota to enter the bloodstream leading to low-grade systemic inflammation. The increased levels of inflammation associated cytokines in circulation has the potential to affect all organs, including the brain. Although the brain is protected by the blood brain barrier, inflammation associated cytokines can damage the junctions in this barrier and allow brain infiltration of peripheral immune cells. Central inflammation in the brain is associated with various neurodegenerative disease such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and neuropsychiatric disorders, namely, depression, and anxiety. Neurodegenerative diseases are of particular concern due to the anticipated rise in the population of the elderly and consequently, the prevalence of these diseases. Additionally, depression and anxiety are the most common mental illnesses affecting roughly 18% of the American population. In this review, we will explore the mechanisms by which IBS can influence the risk and severity of neurological disease.
Collapse
Affiliation(s)
- Yumna El-Hakim
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan, TX, USA
| | - Shameena Bake
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan, TX, USA
| | - Kathiresh Kumar Mani
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan, TX, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan, TX, USA.
| |
Collapse
|
147
|
Crosstalk between Oxidative Stress and Inflammatory Liver Injury in the Pathogenesis of Alcoholic Liver Disease. Int J Mol Sci 2022; 23:ijms23020774. [PMID: 35054960 PMCID: PMC8775426 DOI: 10.3390/ijms23020774] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is characterized by the injury, inflammation, and scarring in the liver owing to excessive alcohol consumption. Currently, ALD is a leading cause for liver transplantation. Therefore, extensive studies (in vitro, in experimental ALD models and in humans) are needed to elucidate pathological features and pathogenic mechanisms underlying ALD. Notably, oxidative changes in the liver have been recognized as a signature trait of ALD. Progression of ALD is linked to the generation of highly reactive free radicals by reactions involving ethanol and its metabolites. Furthermore, hepatic oxidative stress promotes tissue injury and, in turn, stimulates inflammatory responses in the liver, forming a pathological loop that promotes the progression of ALD. Accordingly, accumulating further knowledge on the relationship between oxidative stress and inflammation may help establish a viable therapeutic approach for treating ALD.
Collapse
|
148
|
Singh RP, Niharika J, Kondepudi KK, Bishnoi M, Tingirikari JMR. Recent understanding of human milk oligosaccharides in establishing infant gut microbiome and roles in immune system. Food Res Int 2022; 151:110884. [PMID: 34980411 DOI: 10.1016/j.foodres.2021.110884] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022]
Abstract
Human milk oligosaccharides (HMOs) are complex sugars with distinctive structural diversity present in breast milk. HMOs have various functional roles to play in infant development starting from establishing the gut microbiome and immune system to take it up to the mature phase. It has been a major energy source for human gut microbes that confer positive benefits on infant health by directly interacting through intestinal cells and generating short-chain fatty acids. It has recently become evident that each species of Bifidobacterium and other genera which are resident of the infant gut employ distinct molecular mechanisms to capture and digest diverse structural HMOs to avoid competition among themselves and successfully maintain gut homeostasis. HMOs also directly modulate gut immune responses and can decoy receptors of pathogenic bacteria and viruses, inhibiting their binding on intestinal cells, thus preventing the emergence of a disease. This review provides a critical understanding of how different gut bacteria capture and utilize selective sugars from the HMO pool and how different structural HMOs protect infants from infectious diseases.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Laboratory of Gut Glycobiology, Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS Nagar, Punjab 140306, India.
| | - Jayashree Niharika
- Laboratory of Gut Glycobiology, Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS Nagar, Punjab 140306, India
| | - Kanthi Kiran Kondepudi
- Healthy Gut Research Group, Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS Nagar, Punjab 140306, India
| | - Mahendra Bishnoi
- Healthy Gut Research Group, Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS Nagar, Punjab 140306, India
| | - Jagan Mohan Rao Tingirikari
- Department of Biotechnology, National Institute of Technology Andhra Pradesh, Tadepalligudem, Andhra Pradesh 534101, India
| |
Collapse
|
149
|
Singh RP, Shadan A, Ma Y. Biotechnological Applications of Probiotics: A Multifarious Weapon to Disease and Metabolic Abnormality. Probiotics Antimicrob Proteins 2022; 14:1184-1210. [PMID: 36121610 PMCID: PMC9483357 DOI: 10.1007/s12602-022-09992-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2022] [Indexed: 12/25/2022]
Abstract
Consumption of live microorganisms "Probiotics" for health benefits and well-being is increasing worldwide. Their use as a therapeutic approach to confer health benefits has fascinated humans for centuries; however, its conceptuality gradually evolved with methodological advancement, thereby improving our understanding of probiotics-host interaction. However, the emerging concern regarding safety aspects of live microbial is enhancing the interest in non-viable or microbial cell extracts, as they could reduce the risks of microbial translocation and infection. Due to technical limitations in the production and formulation of traditionally used probiotics, the scientific community has been focusing on discovering new microbes to be used as probiotics. In many scientific studies, probiotics have been shown as potential tools to treat metabolic disorders such as obesity, type-2 diabetes, non-alcoholic fatty liver disease, digestive disorders (e.g., acute and antibiotic-associated diarrhea), and allergic disorders (e.g., eczema) in infants. However, the mechanistic insight of strain-specific probiotic action is still unknown. In the present review, we analyzed the scientific state-of-the-art regarding the mechanisms of probiotic action, its physiological and immuno-modulation on the host, and new direction regarding the development of next-generation probiotics. We discuss the use of recently discovered genetic tools and their applications for engineering the probiotic bacteria for various applications including food, biomedical applications, and other health benefits. Finally, the review addresses the future development of biological techniques in combination with clinical and preclinical studies to explain the molecular mechanism of action, and discover an ideal multifunctional probiotic bacterium.
Collapse
Affiliation(s)
- Rajnish Prakash Singh
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand India
| | - Afreen Shadan
- Dr. Shyama Prasad Mukherjee University, Ranchi, Jharkhand India
| | - Ying Ma
- College of Resource and Environment, Southwest University, Chongqing, China
| |
Collapse
|
150
|
Ping Q, Zhang J, Tang R, Liao S, Zhang Z, Li Y. Effect of surfactants on phosphorus release and acidogenic fermentation of waste activated sludge containing different aluminium phosphate forms. CHEMOSPHERE 2022; 287:132213. [PMID: 34560494 DOI: 10.1016/j.chemosphere.2021.132213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 06/13/2023]
Abstract
The effects of different surfactants (rhamnolipid, trehalolipid and citrate) on phosphorus (P) release and acidogenic fermentation of waste activated sludge (WAS) containing different aluminium phosphate forms (AlPO4, Al(PO3)3) were investigated. Results showed that rhamnolipid was the most effective surfactant to release P from aluminum phosphates (AlPs)-rich sludge. Al(PO3)3 was easier to release P than AlPO4 in WAS due to their different crystal structures. Different surfactants promoted the production of different types of protein. The addition of rhamnolipid was conducive to produce propionate from WAS, while trehalolipid and citrate increased the production of n-butyrate and acetate, respectively. Citrobacter played an important role in producing phosphatase continuously for P release with rhamnolipid addition. Predictive functional profiling indicates that rhamnolipid greatly facilitated adenosine triphosphate (ATP)-binding cassette transporter and quorum sensing. These important discoveries help to enrich P recovery paths from sludge produced with Al-based coagulants in wastewater treatment plants.
Collapse
Affiliation(s)
- Qian Ping
- State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, PR China
| | - Jingyi Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Ruijie Tang
- State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Shuting Liao
- State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Zhipeng Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China
| | - Yongmei Li
- State Key Laboratory of Pollution Control and Resource Reuse, College of Environmental Science and Engineering, Tongji University, Shanghai, 200092, PR China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai, 200092, PR China.
| |
Collapse
|