201
|
Viazis N, Argyriou K, Kotzampassi K, Christodoulou DK, Apostolopoulos P, Georgopoulos SD, Liatsos C, Giouleme O, Koustenis K, Veretanos C, Stogiannou D, Moutzoukis M, Poutakidis C, Mylonas II, Tseti I, Mantzaris GJ. A Four-Probiotics Regimen Combined with A Standard Helicobacter pylori-Eradication Treatment Reduces Side Effects and Increases Eradication Rates. Nutrients 2022; 14:632. [PMID: 35276991 PMCID: PMC8838490 DOI: 10.3390/nu14030632] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/18/2022] Open
Abstract
Aim: To establish whether the addition of probiotics to a globally accepted Helicobacter pylori (H. pylori)-eradication scheme may reduce the rates of side effects and increase the eradication rates. Methods. Prospective, randomized, placebo-controlled trial of patients receiving eradication therapy for H. pylori in the eight participating centers. All patients received a 10-day proton pump inhibitor containing non-bismuth quadruple therapeutic regimen for H. pylori eradication (omeprazole 20 mg, amoxycillin 1 g, clarithromycin 500 mg, and metronidazole 500 mg all twice daily orally) and were randomized to receive either probiotics (group A) or placebo (group B). The probiotic used combined four probiotic strains, i.e., Lactobacillus Acidophilus, Lactiplantibacillus plantarum, Bifidobacterium lactis, and Saccharomyces boulardii. Results. Data were analyzed for 329 patients in group A and 335 patients in group B. Fifty six (17.0%) patients in group A and 170 (50.7%) patients in group B reported the occurrence of an H. pylori treatment-associated new symptom or the aggravation of a pre-existing symptom of any severity (p < 0.00001). H. pylori was successfully eradicated in 303 patients in group A (92.0%) and 291 patients in group B (86.8%), (p = 0.028). Conclusion: Adding probiotics to the 10-day concomitant non-bismuth quadruple H. pylori eradication regimen increases the eradication rate and decreases side effects.
Collapse
Affiliation(s)
- Nikos Viazis
- Gastroenterology Department, Evangelismos Hospital, 10676 Athens, Greece; (K.K.); (C.V.); (G.J.M.)
| | - Konstantinos Argyriou
- Department of Gastroenterology, University Hospital of Larisa, 41334 Larissa, Greece;
| | - Katerina Kotzampassi
- Endoscopy Unit, Department of Surgery, Aristotle University of Thessaloniki, 15341 Athens, Greece; (K.K.); (D.S.)
| | - Dimitrios K. Christodoulou
- Department of Gastroenterology, University Hospital of Ioannina, 45500 Ioannina, Greece; (D.K.C.); (M.M.)
| | - Periklis Apostolopoulos
- Gastroenterology Department, Army Share Fund Hospital (NIMTS), 11521 Athens, Greece; (P.A.); (C.P.)
| | | | - Christos Liatsos
- Gastroenterology Department, 401 General Military Hospital of Athens, 11525 Athens, Greece; (C.L.); (I.I.M.)
| | - Olga Giouleme
- Second Propedeutic Department of Internal Medicine, Medical School, Hippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Kanellos Koustenis
- Gastroenterology Department, Evangelismos Hospital, 10676 Athens, Greece; (K.K.); (C.V.); (G.J.M.)
| | - Christos Veretanos
- Gastroenterology Department, Evangelismos Hospital, 10676 Athens, Greece; (K.K.); (C.V.); (G.J.M.)
| | - Dimitris Stogiannou
- Endoscopy Unit, Department of Surgery, Aristotle University of Thessaloniki, 15341 Athens, Greece; (K.K.); (D.S.)
| | - Miltiadis Moutzoukis
- Department of Gastroenterology, University Hospital of Ioannina, 45500 Ioannina, Greece; (D.K.C.); (M.M.)
| | - Charalambos Poutakidis
- Gastroenterology Department, Army Share Fund Hospital (NIMTS), 11521 Athens, Greece; (P.A.); (C.P.)
| | - Ioannis Ioardanis Mylonas
- Gastroenterology Department, 401 General Military Hospital of Athens, 11525 Athens, Greece; (C.L.); (I.I.M.)
| | - Ioulia Tseti
- Uni-Pharma Kleon Tsetis Pharmaceutical Laboratories S.A., 14564 Athens, Greece;
| | - Gerassimos J. Mantzaris
- Gastroenterology Department, Evangelismos Hospital, 10676 Athens, Greece; (K.K.); (C.V.); (G.J.M.)
| |
Collapse
|
202
|
Araji G, Maamari J, Ahmad FA, Zareef R, Chaftari P, Yeung SCJ. The Emerging Role of the Gut Microbiome in the Cancer Response to Immune Checkpoint Inhibitors: A Narrative Review. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2022; 5:13-25. [PMID: 35663831 PMCID: PMC9138420 DOI: 10.36401/jipo-21-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/10/2021] [Accepted: 09/16/2021] [Indexed: 12/02/2022]
Abstract
The discovery of immune checkpoint inhibitors (ICIs) has revolutionized the care of cancer patients. However, the response to ICI therapy exhibits substantial interindividual variability. Efforts have been directed to identify biomarkers that predict the clinical response to ICIs. In recent years, the gut microbiome has emerged as a critical player that influences the efficacy of immunotherapy. An increasing number of studies have suggested that the baseline composition of a patient's gut microbiota and its dysbiosis are correlated with the outcome of cancer immunotherapy. This review tackles the rapidly growing body of evidence evaluating the relationship between the gut microbiome and the response to ICI therapy. Additionally, this review highlights the impact of antibiotic-induced dysbiosis on ICI efficacy and discusses the possible therapeutic interventions to optimize the gut microbiota composition to augment immunotherapy efficacy.
Collapse
Affiliation(s)
- Ghada Araji
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Julian Maamari
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Fatima Ali Ahmad
- LAU Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Rana Zareef
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Patrick Chaftari
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
203
|
Gregor A, Pignitter M, Trajanoski S, Auernigg-Haselmaier S, Somoza V, König J, Duszka K. Microbial contribution to the caloric restriction-triggered regulation of the intestinal levels of glutathione transferases, taurine, and bile acid. Gut Microbes 2022; 13:1992236. [PMID: 34693866 PMCID: PMC8547879 DOI: 10.1080/19490976.2021.1992236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [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
Recently we showed that caloric restriction (CR) triggers an increase in the levels of free taurine, taurine-conjugated bile acids (BA), and other taurine conjugates in intestinal mucosa while decreasing glutathione (GSH) levels in wild-type male mice. In the current project, we decided to investigate whether the microbiota is involved in the response to CR by depleting gut bacteria. The antibiotics treatment diminished CR-specific increase in the levels of free taurine and its conjugates as well as upregulated expression and activity of GSH transferases (GST) in the intestinal mucosa. Further, it diminished a CR-related increase in BAs levels in the liver, plasma, and intestinal mucosa. Transplant of microbiota from CR mice to ad libitum fed mice triggered CR-like changes in MGST1 expression, levels of taurine and taurine conjugates in the mucosa of the ileum. We show for the first time, that microbiota contributes to the intestinal response to CR-triggered changes in BA, taurine, and GST levels.
Collapse
Affiliation(s)
- András Gregor
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Marc Pignitter
- Department of Physiological Chemistry, University of Vienna, Vienna, Austria
| | - Slave Trajanoski
- Core Facility Computational Bioanalytics, Medical University of Graz, Graz, Austria
| | | | - Veronika Somoza
- Department of Physiological Chemistry, University of Vienna, Vienna, Austria,Leibniz-Institut for Food Systems Biology, Technical University of Munich, Munich, Germany
| | - Jürgen König
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | - Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria,CONTACT Kalina Duszka Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
204
|
Tam RY, van Dorst JM, McKay I, Coffey M, Ooi CY. Intestinal Inflammation and Alterations in the Gut Microbiota in Cystic Fibrosis: A Review of the Current Evidence, Pathophysiology and Future Directions. J Clin Med 2022; 11:649. [PMID: 35160099 PMCID: PMC8836727 DOI: 10.3390/jcm11030649] [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: 12/21/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis (CF) is a life-limiting autosomal recessive multisystem disease. While its burden of morbidity and mortality is classically associated with pulmonary disease, CF also profoundly affects the gastrointestinal (GI) tract. Chronic low-grade inflammation and alterations to the gut microbiota are hallmarks of the CF intestine. The etiology of these manifestations is likely multifactorial, resulting from cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction, a high-fat CF diet, and the use of antibiotics. There may also be a bidirectional pathophysiological link between intestinal inflammation and changes to the gut microbiome. Additionally, a growing body of evidence suggests that these GI manifestations may have significant clinical associations with growth and nutrition, quality of life, and respiratory function in CF. As such, the potential utility of GI therapies and long-term GI outcomes are areas of interest in CF. Further research involving microbial modulation and multi-omics techniques may reveal novel insights. This article provides an overview of the current evidence, pathophysiology, and future research and therapeutic considerations pertaining to intestinal inflammation and alterations in the gut microbiota in CF.
Collapse
Affiliation(s)
- Rachel Y. Tam
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Josie M. van Dorst
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
| | - Isabelle McKay
- Wagga Wagga Base Hospital, Wagga Wagga, NSW 2650, Australia;
| | - Michael Coffey
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| | - Chee Y. Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2031, Australia; (R.Y.T.); (J.M.v.D.); (M.C.)
- Department of Gastroenterology, Sydney Children’s Hospital Randwick, Sydney, NSW 2031, Australia
| |
Collapse
|
205
|
Bai X, Zhu M, He Y, Wang T, Tian D, Shu J. The impacts of probiotics in eradication therapy of Helicobacter pylori. Arch Microbiol 2022; 204:692. [PMID: 36344628 PMCID: PMC9640438 DOI: 10.1007/s00203-022-03314-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/25/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022]
Abstract
Helicobacter pylori (H. pylori) is a well-known pathogen that infects approximately half of the world's population. It is a pathogenic agent with potential health hazards related to diverse diseases, especially digestive diseases, such as chronic gastritis, peptic ulcer, and gastric carcinoma. In clinical, antibiotics are commonly applied in eradication therapy of H. pylori. However, the increase in antibiotic resistance and side effects has induced the failure of eradication therapy. Recent studies have shown that probiotic supplementation has promising application prospects. It can restore the gastrointestinal microbiota balance and prevent dysbacteriosis caused by antibiotics. Furthermore, it has been reported to have direct or indirect inhibitory effects on H. pylori. Probiotics may have a beneficial effect on H. pylori eradication. However, the strain, dosages, duration times, and safety of probiotic supplementation need further study before clinical applications.
Collapse
Affiliation(s)
- Xiaofen Bai
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
- Department of Gastroenterology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Minjie Zhu
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Yajun He
- Department of Clinic Laboratory, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Tengyan Wang
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Da Tian
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Jianchang Shu
- Department of Gastroenterology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
206
|
Gut Dysbiosis Associated with Antibiotics and Disease Severity and Its Relation to Mortality in Critically Ill Patients. Dig Dis Sci 2022; 67:2420-2432. [PMID: 33939152 PMCID: PMC8090918 DOI: 10.1007/s10620-021-07000-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND The gut microbiota are reported to be altered in critical illness. The pattern and impact of dysbiosis on prognosis has not been thoroughly investigated in the ICU setting. AIMS We aimed to evaluate changes in the gut microbiota of ICU patients via 16S rRNA gene deep sequencing, assess the association of the changes with antibiotics use or disease severity, and explore the association of gut microbiota changes with ICU patient prognosis. METHODS Seventy-one mechanically ventilated patients were included. Fecal samples were collected serially on days 1-2, 3-4, 5-7, 8-14, and thereafter when suitable. Microorganisms of the fecal samples were profiled by 16S rRNA gene deep sequencing. RESULTS Proportions of the five major phyla in the feces were diverse in each patient at admission. Those of Bacteroidetes and Firmicutes especially converged and stabilized within the first week from admission with a reduction in α-diversity (p < 0.001). Significant differences occurred in the proportional change of Actinobacteria between the carbapenem and non-carbapenem groups (p = 0.030) and that of Actinobacteria according to initial SOFA score and changes in the SOFA score (p < 0.001). An imbalance in the ratio of Bacteroidetes to Firmicutes within seven days from admission was associated with higher mortality when the ratio was > 8 or < 1/8 (odds ratio: 5.54, 95% CI: 1.39-22.18, p = 0.015). CONCLUSIONS Broad-spectrum antibiotics and disease severity may be associated with gut dysbiosis in the ICU. A progression of dysbiosis occurring in the gut of ICU patients might be associated with mortality.
Collapse
|
207
|
Ou Y, Belzer C, Smidt H, de Weerth C. Development of the gut microbiota in healthy children in the first ten years of life: associations with internalizing and externalizing behavior. Gut Microbes 2022; 14:2038853. [PMID: 35188073 PMCID: PMC8865293 DOI: 10.1080/19490976.2022.2038853] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Increasing evidence indicates that psychopathological disorders are associated with the gut microbiota. However, data are largely lacking from long-term longitudinal birth cohorts, especially those comprising low-risk healthy individuals. Therefore, this study aims to describe gut microbiota development in healthy children from birth till age 10 years, as well as to investigate potential associations with internalizing and externalizing behavior. RESULTS Fecal microbial composition of participants in an ongoing longitudinal study (N = 193) was analyzed at 1, 3 and 4 months, and 6 and 10 years of age by 16S ribosomal RNA gene sequencing. Based on these data, three clusters were identified in infancy, two of which were predominated by Bifidobacterium. In childhood, four clusters were observed, two of which increased in prevalence with age. One of the childhood clusters, similar to an enterotype, was highly enriched in genus-level taxon Prevotella_9. Breastfeeding had marked associations with microbiota composition up till age 10, implying an extended role in shaping gut microbial ecology. Microbial clusters were not associated with behavior. However, Prevotella_9 in childhood was positively related to mother-reported externalizing behavior at age 10; this was validated in child reports. CONCLUSIONS This study validated previous findings on Bifidobacterium-enriched and -depleted clusters in infancy. Importantly, it also mapped continued development of gut microbiota in middle childhood. Novel associations between gut microbial composition in the first 10 years of life (especially Prevotella_9), and externalizing behavior at age 10 were found. Replications in other cohorts, as well as follow-up assessments, will help determine the significance of these findings.
Collapse
Affiliation(s)
- Yangwenshan Ou
- Department of Agrotechnology and Food Sciences, Laboratory of Microbiology, Wageningen University & Research, P.O. Box 8033, EH Wageningen, 6700Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, P.O. Box 9010, 6500GL Nijmegen, Netherlands
| | - Clara Belzer
- Department of Agrotechnology and Food Sciences, Laboratory of Microbiology, Wageningen University & Research, P.O. Box 8033, EH Wageningen, 6700Netherlands
| | - Hauke Smidt
- Department of Agrotechnology and Food Sciences, Laboratory of Microbiology, Wageningen University & Research, P.O. Box 8033, EH Wageningen, 6700Netherlands
| | - Carolina de Weerth
- Donders Institute for Brain, Cognition and Behaviour, Department of Cognitive Neuroscience, Radboud University Medical Center, P.O. Box 9010, 6500GL Nijmegen, Netherlands
| |
Collapse
|
208
|
Xu Y, Zheng S, Jiang S, Chen J, Zhu X, Zhang Y. The effect of Chinese herbal formulas combined with metformin on modulating the gut microbiota in the amelioration of type 2 diabetes mellitus: A systematic review and meta-analysis. Front Endocrinol (Lausanne) 2022; 13:927959. [PMID: 36187136 PMCID: PMC9521410 DOI: 10.3389/fendo.2022.927959] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/15/2022] [Indexed: 12/02/2022] Open
Abstract
UNLABELLED To assess and analyse the effectiveness and safety of combined Chinese herbal formula (CHF) and metformin treatment in the modulation of the gut microbiota in the amelioration of type 2 diabetes mellitus(T2DM), all publications addressing the effect of this combination treatment on the quantitative alterations in the gut microbiota and glucose parameters were collected. Rob tool in the Cochrane handbook was performed to evaluate the methodological quality of all included studies. Relevant information and statistics were abstracted and synthesized in Review Manager 5.4 to evaluate the efficacy of combination treatment. Sensitivity analyses and subgroup analyses were used to analyse the sources of heterogeneity. Publication bias analyses were performed by Stata software to assess the robustness and quality of the outcomes. As a result, a total of 12 eligible RCTs with 1307 T2DM participants from 7 electronic databases were included. Combined CHF with metformin treatment showed better efficacies than metformin monotherapy in regulating the structure of the gut microbiota, characterized by increased Bifidobacterium, Lactobacillus and Bacteroidetes and decreased Enterobacteriaceae, Enterococcus, and Saccharomyces along with better decreases in glycated haemoglobin, fasting plasma glucose, 2-hour postprandial blood glucose, fasting insulin and homeostasis model assessment of insulin resistance. Subgroup analyses further analysed the effect of metformin doses and CHF classifications on controlling hyperglycaemia and altering the gut microbiota. In conclusion, our meta-analysis suggested that combined CHF with metformin treatment is promising for the modulation of the gut microbiota along with ameliorating hyperglycemia in T2DM patients. Importantly, more well-designed RCTs are needed to validate the outcomes and verify the treatment value for clinical purposes. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021291524, identifier CRD42021291524.
Collapse
|
209
|
Tokarek J, Gadzinowska J, Młynarska E, Franczyk B, Rysz J. What Is the Role of Gut Microbiota in Obesity Prevalence? A Few Words about Gut Microbiota and Its Association with Obesity and Related Diseases. Microorganisms 2021; 10:microorganisms10010052. [PMID: 35056501 PMCID: PMC8777962 DOI: 10.3390/microorganisms10010052] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 01/09/2023] Open
Abstract
Obesity is becoming the most dangerous lifestyle disease of our time, and its effects are already being observed in both developed and developing countries. The aim of this study was to investigate the impact of gut microbiota on the prevalence of obesity and associated morbidities, taking into consideration underlying molecular mechanisms. In addition to exploring the relationship between obesity and fecal microorganisms with their metabolites, the study also focused on the factors that would be able to stimulate growth and remodeling of microbiota. Assessed articles were carefully classified according to a predetermined criterion and were critically appraised and used as a basis for conclusions. The considered articles and reviews acknowledge that intestinal microbiota forms a multifunctional system that might significantly affect human homeostasis. It has been proved that alterations in the gut microbiota are found in obese and metabolically diseased patients. The imbalance of microbiome composition, such as changes in Bacteroidetes/Firmicutes ratio and presence of different species of genus Lactobacillus, might promote obesity and comorbidities (type 2 diabetes mellitus, hypertension, dyslipidemia, depression, obstructive sleep apnea). However, there are also studies that contradict this theory. Therefore, further well-designed studies are needed to improve the knowledge about the influence of microbiota, its metabolites, and probiotics on obesity.
Collapse
|
210
|
Clegg TJ, Kawmi N, Graziane NM. Different classes of antibiotics have varying effects on the risk of developing opioid use disorder: a national database study. JOURNAL OF SUBSTANCE USE 2021. [DOI: 10.1080/14659891.2021.2010140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Taylor J. Clegg
- Doctor of Medicine Program, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Noor Kawmi
- Doctor of Medicine Program, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Nicholas M. Graziane
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
211
|
Larussa T, Abenavoli L, Fabiano G, Mancuso MA, Polimeni N, Dumitrascu DL, Luzza F. Gut microbiota in inflammatory bowel disease: a target for therapy not to be missed. Minerva Gastroenterol (Torino) 2021; 67:357-368. [PMID: 35040302 DOI: 10.23736/s2724-5985.21.02907-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last years, the gut microbiota achieved great importance, since several studies demonstrated its correlation with the immune system and with the maintenance of intestinal homeostasis, as well as with the regulation of the integrity of the epithelium and the intestinal motility. An imbalance in microbial species promotes a dysbiosis, which has been associated with chronic diseases such as metabolic syndrome, inflammatory diseases, and some behavior disorders. The association with gut microbiota and dysbiosis has been demonstrated mostly in inflammatory bowel disease (IBD). Several studies investigated the application of antibiotics, prebiotics, probiotics, and fecal microbiota transplantation in the treatment strategies for IBD. In this review, we discuss the recent findings on the potential role of the gut microbiota manipulation, with particular attention to bacterial microbiota, which could be implicated for a successful IBD therapeutic approach.
Collapse
Affiliation(s)
- Tiziana Larussa
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy -
| | - Ludovico Abenavoli
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Giulia Fabiano
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Maria A Mancuso
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| | - Natale Polimeni
- Digestive Endoscopy Service, Casa di Cura Policlinico Madonna della Consolazione, Reggio Calabria, Italy
| | - Dan L Dumitrascu
- Second Medical Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Francesco Luzza
- Department of Health Sciences, Magna Græcia University, Catanzaro, Italy
| |
Collapse
|
212
|
Huang X, Li M, Hou S, Tian B. Role of the microbiome in systemic therapy for pancreatic ductal adenocarcinoma (Review). Int J Oncol 2021; 59:101. [PMID: 34738624 PMCID: PMC8577795 DOI: 10.3892/ijo.2021.5281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
A large body of evidence has revealed that the microbiome serves a role in all aspects of cancer, particularly cancer treatment. To date, studies investigating the relationship between the microbiome and systemic therapy for pancreatic ductal adenocarcinoma (PDAC) are lacking. PDAC is a high‑mortality malignancy (5‑year survival rate; <9% for all stages). Systemic therapy is one of the most important treatment choices for all patients; however, resistance or toxicity can affect its efficacy. Studies have supported the hypothesis that the microbiome is closely associated with the response to systemic therapy in PDAC, including the induction of drug resistance, or toxicity and therapy‑related changes in microbiota composition. The present review comprehensively summarized the role of the microbiome in systemic therapy for PDAC and the associated molecular mechanisms in an attempt to provide a novel direction for the improvement of treatment response and proposed potential directions for in‑depth research.
Collapse
Affiliation(s)
| | | | - Shengzhong Hou
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bole Tian
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
213
|
Yang C, Liang L, Lv P, Liu L, Wang S, Wang Z, Chen Y. Effects of non-viable Lactobacillus reuteri combining with 14-day standard triple therapy on Helicobacter pylori eradication: A randomized double-blind placebo-controlled trial. Helicobacter 2021; 26:e12856. [PMID: 34628695 DOI: 10.1111/hel.12856] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Viable probiotics have shown effects on the eradication of Helicobacter pylori, but the role of non-viable probiotics in H. pylori eradication is unclear. This study aimed to evaluate the effects of non-viable Lactobacillus reuteri DSM17648 combining with 14-day standard triple therapy on H. pylori eradication. MATERIALS AND METHODS Two hundred treatment-naive H. pylori-positive adult patients were randomized equally to receive non-viable L. reuteri DSM17648 (LR group) or placebo for 4 weeks, with the latter 2 weeks treated together with triple therapy. The Gastrointestinal Symptom Rating Scale (GSRS) was completed before and after treatment. Stool samples were collected for 16S rRNA gene sequencing at week0, week2, and week8. RESULTS Eradication rates in the LR group and the placebo group were 81.8% and 83.7% in ITT analysis (p = 0.730), 86.2% and 87.2% in PP analysis (p = 0.830), respectively. After treatment, the mean GSRS score decreased significantly in the LR group as compared with the placebo group (1.9 ± 0.2 vs. 2.7 ± 0.3; p = 0.030). Significantly less patients in the LR group as compared with the placebo group reported abdominal distention (5.1% vs. 16.3%; p = 0.010) and diarrhea (11.1% vs. 23.5%; p = 0.022). The relative abundance of Proteobacteria phylum and Escherichia-Shigella genus in the placebo group was about 4.0-fold and 8.1-fold of that in the LR group at wk2, respectively. Significant changes of diversity and enhancements of Fusicatenibacter, Subdoligranulum, and Faecalibacterium were observed in the LR group compared with the placebo group. CONCLUSIONS Supplementation of non-viable L. reuteri DSM17648 with triple therapy did not improve the eradication rate of H. pylori, but it helped to build up a beneficial microbial profile and reduced the frequencies of abdominal distention, diarrhea, and the GSRS score.
Collapse
Affiliation(s)
- Chenghai Yang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liping Liang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pinjing Lv
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Le Liu
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Siqi Wang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiqing Wang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
214
|
Effects of konjac glucomannan with different molecular weights on gut microflora with antibiotic perturbance in in vitro fecal fermentation. Carbohydr Polym 2021; 273:118546. [PMID: 34560958 DOI: 10.1016/j.carbpol.2021.118546] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/08/2021] [Accepted: 08/08/2021] [Indexed: 02/08/2023]
Abstract
This study investigated the effect of konjac glucomannan (KGM) of different molecular weight on fecal microflora against antibiotic disturbance. KGM (~1.8 × 107 Da) was partially hydrolysed with trifluoroacetic acid (TFA) for 10 and 60 min to KGM1 (~2.1 × 104 Da) and KGM2 (7413 Da), respectively. The acid treatment caused significant reduction of intrinsic viscosity, average molecular weight (MW) and particle size of KGM, but brought limited change to the molecular structure. Low-MW KGM2 showed the most significant effect on fecal microflora in the presence of two common antibiotics (ampicillin and clindamycin), by increasing the relative abundance of Bifidobacteriaceae while decreasing the proportion of Enterobacteriaceae. Additionally, both the native and acid-treated KGM counteracted the adverse influence of antibiotics on the production of short chain fatty acids. The results have demonstrated the effect of KGM on gut microbiota with antibiotic disturbance.
Collapse
|
215
|
Chen Y, Cui W, Li X, Yang H. Interaction Between Commensal Bacteria, Immune Response and the Intestinal Barrier in Inflammatory Bowel Disease. Front Immunol 2021; 12:761981. [PMID: 34858414 PMCID: PMC8632219 DOI: 10.3389/fimmu.2021.761981] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
In inflammatory bowel disease (IBD), intestinal mucosa cell and intestinal epithelial cell are severely damaged, and then their susceptibility to bacteria increases, so many commensal bacteria become pathogenic. The pathogenic commensal bacteria can stimulate a series of compensatory immune responses in the intestine. However, the immune response prevents the intestinal tract from restoring homeostasis, which in turn produces an indispensable inflammatory response. On the contrary, in IBD, the fierce inflammatory response contributes to the development of IBD. However, the effect of commensal bacteria on inflammation in IBD has not been clearly studied. Therefore, we further summarize the changes brought about by the changes of commensal bacteria to the inflammation of the intestines and their mutual influence. This article reviews the protective mechanism of commensal bacteria in healthy people and the mechanism of commensal bacteria and immune response to the destruction of the intestinal barrier when IBD occurs. The treatment and prevention of IBD are also briefly summarized.
Collapse
Affiliation(s)
| | | | - Xiao Li
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
216
|
Aparecida Dos Reis Louzano S, de Moura E Dias M, Lopes da Conceição L, Antônio de Oliveira Mendes T, Gouveia Peluzio MDC. Ceftriaxone causes dysbiosis and changes intestinal structure in adjuvant obesity treatment. Pharmacol Rep 2021; 74:111-123. [PMID: 34757518 DOI: 10.1007/s43440-021-00336-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND Obesity is still a worldwide public health problem, requiring the development of adjuvant therapies to combat it. In this context, modulation of the intestinal microbiota seems prominent, given that the composition of the intestinal microbiota contributes to the outcome of this disease. The aim of this work is to investigate the treatment with an antimicrobial and/or a potential probiotic against overweight. METHODS Male C57BL/6J mice were subjected to a 12-week overweight induction protocol. After that, 4-week treatment was started, with mice divided into four groups: control, treated with distilled water; potential probiotic, with Lactobacillus gasseri LG-G12; antimicrobial, with ceftriaxone; and antimicrobial + potential probiotic with ceftriaxone in the first 2 weeks and L. gasseri LG-G12 in the subsequent weeks. RESULTS The treatment with ceftriaxone in isolated form or in combination with the potential probiotic provided a reduction in body fat. However, such effect is supposed to be a consequence of the negative action of ceftriaxone on the intestinal microbiota composition, and this intestinal dysbiosis may have contributed to the destruction of the intestinal villi structure, which led to a reduction in the absorptive surface. Also, the effects of L. gasseri LG-G12 apparently have been masked by the consumption of the high-fat diet. CONCLUSIONS The results indicate that the use of a ceftriaxone in the adjuvant treatment of overweight is not recommended due to the potential risk of developing inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Mariana de Moura E Dias
- Department of Nutrition and Health, Universidade Federal de Viçosa, Avenue PH Rolfs s/n, Vicosa, MG, 36570-900, Brazil.
| | - Lisiane Lopes da Conceição
- Department of Nutrition and Health, Universidade Federal de Viçosa, Avenue PH Rolfs s/n, Vicosa, MG, 36570-900, Brazil
| | - Tiago Antônio de Oliveira Mendes
- Department of Biochemistry and Molecular Biology, Universidade Federal de Viçosa, Avenue PH Rolfs s/n, Vicosa, MG, 36570-900, Brazil
| | | |
Collapse
|
217
|
Abstract
Lyme disease, which is caused by the spirochete Borrelia burgdorferi, is on the rise. Current treatment relies on broad-spectrum antibiotics that perturb the gut microbiome. In a recent paper in Cell, Leimer et al. demonstrate the utility of a long-forgotten antibiotic, Hygromycin A, as a spirochete-specific antibacterial that is conducive to gut health.
Collapse
|
218
|
Fan P, Kim M, Liu G, Zhai Y, Liu T, Driver JD, Jeong KC. The Gut Microbiota of Newborn Calves and Influence of Potential Probiotics on Reducing Diarrheic Disease by Inhibition of Pathogen Colonization. Front Microbiol 2021; 12:772863. [PMID: 34745079 PMCID: PMC8567051 DOI: 10.3389/fmicb.2021.772863] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 01/17/2023] Open
Abstract
Calf diarrhea is one of the most concerning challenges facing both the dairy and beef cattle industry. Maintaining healthy gut microbiota is essential for preventing gastrointestinal disorders. Here, we observed significantly less bacterial richness in the abnormal feces with watery or hemorrhagic morphology compared to the normal solid feces. The normal solid feces showed high relative abundances of Osllospiraceae, Christensenellaceae, Barnesiella, and Lactobacillus, while the abnormal feces contained more bacterial taxa of Negativicutes, Tyzzerella, Parasutterella, Veillonella, Fusobacterium, and Campylobacter. Healthy calves had extensive bacterial-bacterial correlations, with negative correlation between Lactobacillus and potential diarrheagenic Escherichia coli-Shigella, but not in the abnormal feces. We isolated Lactobacillus species (L. reuteri, L. johnsonii, L. amylovorus, and L. animalis), with L. reuteri being the most abundant, from the healthy gut microbiota. Isolated Lactobacillus strains inhibited pathogenic strains including E. coli K88 and Salmonella Typhimurium. These findings indicate the importance of a diverse gut microbiota in newborn calf’s health and provide multiple potential probiotics that suppress pathogen colonization in the gastrointestinal tract to prevent calf diarrhea.
Collapse
Affiliation(s)
- Peixin Fan
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Miju Kim
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Grace Liu
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Yuting Zhai
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Ting Liu
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Joseph Danny Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Kwangcheol C Jeong
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
219
|
Zhou J, Sun S, Luan S, Xiao X, Yang Y, Mao C, Chen L, Zeng X, Zhang Y, Yuan Y. Gut Microbiota for Esophageal Cancer: Role in Carcinogenesis and Clinical Implications. Front Oncol 2021; 11:717242. [PMID: 34733778 PMCID: PMC8558403 DOI: 10.3389/fonc.2021.717242] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/24/2021] [Indexed: 02/05/2023] Open
Abstract
Esophageal cancer (EC) is a common malignant tumor of the upper digestive tract. The microbiota in the digestive tract epithelium comprises a large number of microorganisms that adapt to the immune defense and interact with the host to form symbiotic networks, which affect many physiological processes such as metabolism, tissue development, and immune response. Reports indicate that there are microbial compositional changes in patients with EC, which provides an important opportunity to advance clinical applications based on findings on the gut microbiota. For example, microbiota detection can be used as a biomarker for screening and prognosis, and microorganism levels can be adjusted to treat cancer and decrease the adverse effects of treatment. This review aims to provide an outline of the gut microbiota in esophageal neoplasia, including the mechanisms involved in microbiota-related carcinogenesis and the prospect of utilizing the microbiota as EC biomarkers and treatment targets. These findings have important implications for translating the use of gut microbiota in clinical applications.
Collapse
Affiliation(s)
- Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shangwei Sun
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Luan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chengyi Mao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
220
|
Li H, Xiang Y, Zhu Z, Wang W, Jiang Z, Zhao M, Cheng S, Pan F, Liu D, Ho RCM, Ho CSH. Rifaximin-mediated gut microbiota regulation modulates the function of microglia and protects against CUMS-induced depression-like behaviors in adolescent rat. J Neuroinflammation 2021; 18:254. [PMID: 34736493 PMCID: PMC8567657 DOI: 10.1186/s12974-021-02303-y] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chronic unpredictable mild stress (CUMS) can not only lead to depression-like behavior but also change the composition of the gut microbiome. Regulating the gut microbiome can have an antidepressant effect, but the mechanism by which it improves depressive symptoms is not clear. Short-chain fatty acids (SCFAs) are small molecular compounds produced by the fermentation of non-digestible carbohydrates. SFCAs are ubiquitous in intestinal endocrine and immune cells, making them important mediators of gut microbiome-regulated body functions. The balance between the pro- and anti-inflammatory microglia plays an important role in the occurrence and treatment of depression caused by chronic stress. Non-absorbable antibiotic rifaximin can regulate the structure of the gut microbiome. We hypothesized that rifaximin protects against stress-induced inflammation and depression-like behaviors by regulating the abundance of fecal microbial metabolites and the microglial functions. METHODS We administered 150 mg/kg rifaximin intragastrically to rats exposed to CUMS for 4 weeks and investigated the composition of the fecal microbiome, the content of short-chain fatty acids in the serum and brain, the functional profiles of microglia and hippocampal neurogenesis. RESULTS Our results show that rifaximin ameliorated depressive-like behavior induced by CUMS, as reflected by sucrose preference, the open field test and the Morris water maze. Rifaximin increased the relative abundance of Ruminococcaceae and Lachnospiraceae, which were significantly positively correlated with the high level of butyrate in the brain. Rifaximin increased the content of anti-inflammatory factors released by microglia, and prevented the neurogenic abnormalities caused by CUMS. CONCLUSIONS These results suggest that rifaximin can regulate the inflammatory function of microglia and play a protective role in pubertal neurodevelopment during CUMS by regulating the gut microbiome and short-chain fatty acids.
Collapse
Affiliation(s)
- Haonan Li
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yujiao Xiang
- Cheeloo Hospital, Shandong University, Jinan, People's Republic of China
| | - Zemeng Zhu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Wei Wang
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Zhijun Jiang
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Mingyue Zhao
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Shuyue Cheng
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Fang Pan
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China.
| | - Roger C M Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| | - Cyrus S H Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
221
|
El-Salhy M, Hausken T, Hatlebakk JG. Current status of fecal microbiota transplantation for irritable bowel syndrome. Neurogastroenterol Motil 2021; 33:e14157. [PMID: 34236740 DOI: 10.1111/nmo.14157] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a common gastrointestinal functional disorder. Although IBS is a benign condition, it reduces the quality of life considerably. While there is currently no effective treatment for this disorder, fecal microbiota transplantation (FMT) seems to be promising. PURPOSE The aim of this review was to analysis possible factors affecting the success or failure of the randomized controlled trials (RCTs) of FMT for IBS and highlighting the gaps in our knowledge that need to be filled and of sketching a possible model for successful FMT in IBS patients. METHODS A systematic search was conducted of literature published in English from January 2015 to December 2020 using the keywords: fecal microbiota transplantation, randomized trials, and IBS. KEY RESULTS Seven randomized controlled trials (RCTs) on the efficacy of FMT for IBS were found in the literature. Four of the seven RCTs found various positive effects, while the other three did not find any effect. CONCLUSIONS AND INFERENCES The efficacy of FMT for IBS appears to be donor-dependent. The effective (super) donor would need to have a favorable microbiota signature, and 11 clinical criteria that are known to be associated with a favorable microbiota have been suggested for selecting FMT donors for IBS. Comparing the microbiota of the effective donors with those of healthy subjects would reveal the favorable microbiota signature required for a super-donor. However, the studies reviewed were not designed to compare efficacy of different donor types. The dose of the fecal transplant is also an important factor influencing the outcome of FMT for IBS. However, further studies designed to test the effect of fecal transplant dose are needed to answer this question. Administering the fecal transplant to either the small or large intestine seems to be effective, but the optimal route of administration remains to be determined. Moreover, whether single or repeated FMT is more effective is also still unclear. A 1-year follow-up of IBS patients who received FMT showed that adverse events of abdominal pain, diarrhea, and constipation were both mild and self-limiting.
Collapse
Affiliation(s)
- Magdy El-Salhy
- Department of Medicine, Stord Hospital, Stord, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, National Centre for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| | - Trygve Hausken
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, National Centre for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| | - Jan Gunnar Hatlebakk
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, National Centre for Functional Gastrointestinal Disorders, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
222
|
Jones MP, Shah A, Walker MM, Koloski NA, Holtmann G, Talley NJ. Antibiotic use but not gastrointestinal infection frequently precedes first diagnosis of functional gastrointestinal disorders. United European Gastroenterol J 2021; 9:1074-1080. [PMID: 34653313 PMCID: PMC8598965 DOI: 10.1002/ueg2.12164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION While the etiopathogenesis of functional gastrointestinal disorders (FGIDs) is not completely understood, alterations of the intestinal microbiome have been observed. Antibiotics can induce dysbiosis, but whether antibiotics are a risk factor for the onset of FGIDs is uncertain. Antibiotics have been reported as both a risk factor for new onset FGID but also as a therapy for existing FGID. This study aimed to estimate the fraction of cases where antibiotics provoked the onset of FGID. METHOD Electronic medical records were obtained from general practices (primary care) in the United Kingdom. Dates of antibiotic prescription (AP) were compared with first date of FGID diagnosis and contrasted across three prevalent FGIDs and controls without gastrointestinal disorders. RESULTS There were 10,926 GI healthy controls, 4326 IBS alone, 3477 FD alone, 340 chronic constipation and 4402 with overlap of multiple conditions. Both the prevalence of AP and rate were higher in FGID patients and increased with diagnosis of multiple FGIDs. 7%-14% of FGID patients were prescribed their first recorded antibiotic in the 12 months prior to their first FGID diagnosis and 20%-33% were prescribed an antibiotic in the same period. Differences between FGID groups were not accounted for by social deprivation and only rate of AP was moderated by social deprivation. In contrast, only 5%-10% of patients ever had a gastrointestinal infection recorded and only 1.5%-3.5% prior to their first FGID diagnosis. CONCLUSION These data indicate that antibiotics are prescribed prior to FGID diagnosis in a significant minority of care-seeking FGID patients, opening the potential for this medication to contribute to the pathophysiology. APs appears to mostly be for non-gastrointestinal conditions.
Collapse
Affiliation(s)
- Michael P. Jones
- School of Psychological SciencesMacquarie UniversityNorth RydeNew South WalesAustralia
| | - Ayesha Shah
- Department of Gastroenterology and HepatologyPrincess Alexandra Hospital and Translational Research Institute (TRI)WoolloongabbaQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Marjorie M. Walker
- College of Health, Medicine and Well BeingUniversity of NewcastleNewcastleNew South WalesAustralia
| | - Natasha A. Koloski
- Department of Gastroenterology and HepatologyPrincess Alexandra Hospital and Translational Research Institute (TRI)WoolloongabbaQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
- College of Health, Medicine and Well BeingUniversity of NewcastleNewcastleNew South WalesAustralia
| | - Gerald Holtmann
- Department of Gastroenterology and HepatologyPrincess Alexandra Hospital and Translational Research Institute (TRI)WoolloongabbaQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Nicholas J. Talley
- College of Health, Medicine and Well BeingUniversity of NewcastleNewcastleNew South WalesAustralia
| |
Collapse
|
223
|
Neroni B, Evangelisti M, Radocchia G, Di Nardo G, Pantanella F, Villa MP, Schippa S. Relationship between sleep disorders and gut dysbiosis: what affects what? Sleep Med 2021; 87:1-7. [PMID: 34479058 DOI: 10.1016/j.sleep.2021.08.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022]
Abstract
Sleep plays a fundamental role in maintaining good psycho-physical health, it can influence hormone levels, mood, and weight. Recent studies, focused on the interconnection between intestinal microbiome and sleep disorders, have shown the growing importance of a healthy and balanced intestinal microbiome for the hosts health. Normally, gut microbiota and his host are linked by mutualistic relationship, that in some conditions, can be compromised by shifts in microbiota's composition, called dysbiosis. Both sleep problems and dysbiosis of the gut microbiome can lead to metabolic disorders and, in this review, we will explore what is present in literature on the link between sleep pathologies and intestinal dysbiosis.
Collapse
Affiliation(s)
- Bruna Neroni
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy
| | | | - Giulia Radocchia
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy
| | - Giovanni Di Nardo
- Sant'Andrea Hospital, NESMOS Department, Sapienza University of Rome, Italy
| | - Fabrizio Pantanella
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy
| | - Maria Pia Villa
- Sant'Andrea Hospital, NESMOS Department, Sapienza University of Rome, Italy
| | - Serena Schippa
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy.
| |
Collapse
|
224
|
Yan W, Zhou Q, Yuan Z, Fu L, Wen C, Yang N, Sun C. Impact of the gut microecology on Campylobacter presence revealed by comparisons of the gut microbiota from chickens raised on litter or in individual cages. BMC Microbiol 2021; 21:290. [PMID: 34686130 PMCID: PMC8532315 DOI: 10.1186/s12866-021-02353-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/11/2021] [Indexed: 11/10/2022] Open
Abstract
Background Poultry is the major reservoir of Campylobacter that contributes to human campylobacteriosis and threatens food safety. Litter contact has been linked to Campylobacter colonization, but the gut microecological impact underlying this link remains not fully clear. Here, we sought to investigate the impact of the gut microecology on the presence of Campylobacter by examining the microbiota in the duodenum, jejunum, ileum, ceca, and feces from chickens raised on commercial litter and in individual cages at 0–57 days of age. Results Through litter contact, the presence of Campylobacter was found to benefit from microecological competition among Lactobacillus, Helicobacter, and genera that are halotolerant and aerobic or facultatively anaerobic in the upper intestine, such as Corynebacterium and Brachybacterium. The presence was also promoted by the increased abundance in obligate anaerobic fermentation microbes, especially members of the orders Clostridiales and Bacteroidales. The longitudinal analysis supported the vertical or pseudo-vertical transmission but suggested that colonization might occur immensely at 7–28 days of age. We observed a host genetic effect on the gut microecology, which might lead to increased heterogeneity of the microecological impact on Campylobacter colonization. Conclusions The findings advance the understanding of the gut microecological impact on Campylobacter presence in the chicken gut under conditions of litter contact and suggest that manipulations of the gut microecology, as well as the microbes identified in the Campylobacter association networks, might be important for the development of intervention strategies. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02353-5.
Collapse
Affiliation(s)
- Wei Yan
- Poultry Science Laboratory, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Qianqian Zhou
- Poultry Science Laboratory, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Zhongyang Yuan
- Poultry Science Laboratory, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Liang Fu
- Poultry Science Laboratory, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Chaoliang Wen
- Poultry Science Laboratory, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Ning Yang
- Poultry Science Laboratory, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.,National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China
| | - Congjiao Sun
- Poultry Science Laboratory, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China. .,National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Beijing, 100193, China.
| |
Collapse
|
225
|
Tong Q, Cui LY, Bie J, Han XY, Hu ZF, Wang HB, Zhang JT. Changes in the gut microbiota diversity of brown frogs (Rana dybowskii) after an antibiotic bath. BMC Vet Res 2021; 17:333. [PMID: 34674716 PMCID: PMC8529755 DOI: 10.1186/s12917-021-03044-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/04/2021] [Indexed: 12/17/2022] Open
Abstract
Background Captive amphibians frequently receive antibiotic baths to control bacterial diseases. The potential collateral effect of these antibiotics on the microbiota of frogs is largely unknown. To date, studies have mainly relied on oral administration to examine the effects of antibiotics on the gut microbiota; in contrast, little is known regarding the effects of bath-applied antibiotics on the gut microbiota. The gut microbiota compositions of the gentamicin, recovery, and control groups were compared by Illumina high-throughput sequencing, and the functional profiles were analysed using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). Furthermore, the relationship between the structure and predicted functional composition of the gut microbiota was determined. Results The alpha diversity indices were significantly reduced by the gentamicin bath, illustrating that this treatment significantly changed the composition of the gut microbiota. After 7 days, the gut microbiota of the recovery group was not significantly different from that of the gentamicin group. Forty-four indicator taxa were selected at the genus level, comprising 42 indicators representing the control group and 2 indicators representing the gentamicin and recovery groups. Potential pathogenic bacteria of the genera Aeromonas, Citrobacter, and Chryseobacterium were significantly depleted after the gentamicin bath. There was no significant positive association between the community composition and functional composition of the gut microbiota in the gentamicin or control frogs, indicating that the functional redundancy of the gut bacterial community was high. Conclusions Gentamicin significantly changed the structure of the gut microbiota of R. dybowskii, and the gut microbiota exhibited weak resilience. However, the gentamicin bath did not change the functional composition of the gut microbiota of R. dybowskii, and there was no significant correlation between the structural composition and the functional composition of the gut microbiota. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-021-03044-z.
Collapse
Affiliation(s)
- Qing Tong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.,Jiamusi Branch of Heilongjiang Academy of Forestry Sciences, Jiamusi, 154002, China.,College of Life Science, Jiamusi University, Jiamusi, 154007, China
| | - Li-Yong Cui
- Jiamusi Branch of Heilongjiang Academy of Forestry Sciences, Jiamusi, 154002, China
| | - Jia Bie
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiao-Yun Han
- College of Life Science, Jiamusi University, Jiamusi, 154007, China
| | - Zong-Fu Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hong-Bin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jian-Tao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
226
|
Khan S, Sharaf M, Ahmed I, Khan TU, Shabana S, Arif M, Kazmi SSUH, Liu C. Potential utility of nano-based treatment approaches to address the risk of Helicobacter pylori. Expert Rev Anti Infect Ther 2021; 20:407-424. [PMID: 34658307 DOI: 10.1080/14787210.2022.1990041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Helicobacter pylori (H. pylori) has occupied a significant place among infectious pathogens and it has been documented as a leading challenge due to its higher resistance to the commonly used drugs, higher adaptability, and lower targeting specificity of the available drugs. AREAS COVERED New treatment strategies are urgently needed in order to improve the current advancement in modern medicine. Nanocarriers have gained an advantage of drug encapsulation and high retention time in the stomach with a prolonged drug release rate at the targeted site. This article aims to highlight the recent advances in nanotechnology with special emphasis on metallic, polymeric, lipid, membrane coated, and target-specific nanoparticles (NPs), as well as, natural products for treating H. pylori infection. We discussed a comprehensive approach to understand H. pylori infection and elicits to rethink about the increasing threat posed by H. pylori and its treatment strategies. EXPERT OPINION To address these issues, nanotechnology has got huge potential to combat H. pylori infection and has made great progress in the field of biomedicine. Moreover, combinatory studies of natural products and probiotics in conjugation with NPs have proven efficiency against H. pylori infection, with an advantage of lower cytotoxicity, minimal side effects, and stronger antibacterial potential.[Figure: see text].
Collapse
Affiliation(s)
- Sohaib Khan
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Mohamed Sharaf
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao, China.,Department of Biochemistry, Faculty of Agriculture, AL-Azhar University, Nasr City, Egypt
| | | | | | - Samah Shabana
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Muhammad Arif
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | | | - Chenguang Liu
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| |
Collapse
|
227
|
Mishra K, Bukavina L, Ghannoum M. Symbiosis and Dysbiosis of the Human Mycobiome. Front Microbiol 2021; 12:636131. [PMID: 34630340 PMCID: PMC8493257 DOI: 10.3389/fmicb.2021.636131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
The influence of microbiological species has gained increased visibility and traction in the medical domain with major revelations about the role of bacteria on symbiosis and dysbiosis. A large reason for these revelations can be attributed to advances in deep-sequencing technologies. However, the research on the role of fungi has lagged. With the continued utilization of sequencing technologies in conjunction with traditional culture assays, we have the opportunity to shed light on the complex interplay between the bacteriome and the mycobiome as they relate to human health. In this review, we aim to offer a comprehensive overview of the human mycobiome in healthy and diseased states in a systematic way. The authors hope that the reader will utilize this review as a scaffolding to formulate their understanding of the mycobiome and pursue further research.
Collapse
Affiliation(s)
- Kirtishri Mishra
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, OH, United States.,Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Laura Bukavina
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, OH, United States.,Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Mahmoud Ghannoum
- Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Center for Medical Mycology, and Integrated Microbiome Core, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Dermatology, Case Western Reserve University School of Medicine and University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
228
|
Ding M, Lang Y, Shu H, Shao J, Cui L. Microbiota-Gut-Brain Axis and Epilepsy: A Review on Mechanisms and Potential Therapeutics. Front Immunol 2021; 12:742449. [PMID: 34707612 PMCID: PMC8542678 DOI: 10.3389/fimmu.2021.742449] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022] Open
Abstract
The gut-brain axis refers to the bidirectional communication between the gut and brain, and regulates intestinal homeostasis and the central nervous system via neural networks and neuroendocrine, immune, and inflammatory pathways. The development of sequencing technology has evidenced the key regulatory role of the gut microbiota in several neurological disorders, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Epilepsy is a complex disease with multiple risk factors that affect more than 50 million people worldwide; nearly 30% of patients with epilepsy cannot be controlled with drugs. Interestingly, patients with inflammatory bowel disease are more susceptible to epilepsy, and a ketogenic diet is an effective treatment for patients with intractable epilepsy. Based on these clinical facts, the role of the microbiome and the gut-brain axis in epilepsy cannot be ignored. In this review, we discuss the relationship between the gut microbiota and epilepsy, summarize the possible pathogenic mechanisms of epilepsy from the perspective of the microbiota gut-brain axis, and discuss novel therapies targeting the gut microbiota. A better understanding of the role of the microbiota in the gut-brain axis, especially the intestinal one, would help investigate the mechanism, diagnosis, prognosis evaluation, and treatment of intractable epilepsy.
Collapse
Affiliation(s)
| | | | | | | | - Li Cui
- Department of Neurology, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
229
|
Liu G, Guo Z, Liu D, Meng H, Zheng Y, Zhao X, Gu L, Chen Z, Chen X, Li M, Sun J, Ma Z, He H, Yu X, Hu F. Does gut microbiota regulate brooding in geese? ANIM BIOL 2021. [DOI: 10.1163/15707563-bja10059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abstract
Domestic geese can reduce the amount of food intake when brooding. Because of the reduction in food intake, the total number of microorganisms in the gut is also reduced. Will this affect the goose’s thinking and make the goose stop brooding and eat food? We hypothesize that gut microbiota affects the brain through a brain–gut peptide and further regulates the breeding behavior of geese. In this study, we evaluated the microbiome related to the goose and transcription groups of brooding and egg production periods. The changes and differences in gut microbiota and gene expression of female geese in different reproduction periods were analyzed, and the possible interaction between them was explored. The results showed that the relative abundance of Faecalibacterium with a growth-promoting effect in the cecum was higher in the egg production group than in the brooding group. Microbial metabolic pathways with significant differences between the two groups were also enriched in the secondary functional groups with different gut microbiota metabolism. The downregulated genes in the egg production group were mainly related to energy metabolism, such as ATP synthesis-related genes. These results suggest that the brooding group’s gut microbiota can make relevant changes according to the reproduction stage of the goose. Since the amount of food taken in is reduced, it can promote the decomposition of the host’s fat. Simultaneously, insulin is also used to deliver messages to the brain; it is necessary to end the brooding behavior at an appropriate time and for eating to start.
Collapse
Affiliation(s)
- Guojun Liu
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - Zhenhua Guo
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - Di Liu
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - He Meng
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University; Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 200240, P.R. China
| | - Yuming Zheng
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University; Shanghai Key Laboratory of Veterinary Biotechnology, Shanghai 200240, P.R. China
| | - Xiuhua Zhao
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - Lihong Gu
- Hainan Academy of Agricultural Sciences, Institute of Animal Science and Veterinary Medicine, 14 Xingdan Road, Qiongshan District, Haikou, 570203, P.R. China
| | - Zhifeng Chen
- Heilongjiang Academy of Agricultural Sciences, Qiqihare Branch Academy, No. 2 Heyi Road, Qiqihare 161005, P.R. China
| | - Xingyong Chen
- College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiangxi Road, Hefei, 230036, P.R. China
| | - Manyu Li
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - Jinyan Sun
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - Zhigang Ma
- Heilongjiang Academy of Agricultural Sciences, Qiqihare Branch Academy, No. 2 Heyi Road, Qiqihare 161005, P.R. China
| | - Haijuan He
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - Xiaolong Yu
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, P.R. China
| | - Fanghong Hu
- Agricultural and Rural Bureau, Longhexi Road, Liuan, 237006, P.R. China
| |
Collapse
|
230
|
Maity C, Gupta AK. Therapeutic efficacy of probiotic Alkalihalobacillus clausii 088AE in antibiotic-associated diarrhea: A randomized controlled trial. Heliyon 2021; 7:e07993. [PMID: 34585011 PMCID: PMC8453216 DOI: 10.1016/j.heliyon.2021.e07993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/26/2021] [Accepted: 09/10/2021] [Indexed: 01/22/2023] Open
Abstract
The use of probiotics in gastrointestinal ailments has shown therapeutic effects. The imbalance of the microbiota caused by antibiotic treatment or others has been shown to be restored to normal with probiotic treatment. In this study, a genomically and phenotypically safe probiotic Alkalihalobacillus clausii 088AE has been evaluated for ameliorating antibiotic-associated diarrhea (AAD) in pediatrics (PE, n = 60, 2-10 years), adolescent and adults (AA, n = 60, 11-65 years) through a randomized controlled clinical trial. A. clausii 088AE was administered for seven days (PE, 4 and AA, 6 billion/day) and primary and secondary endpoints were evaluated on different visits. Compared to the respective placebo arms, A. clausii 088AE improved the diarrheal (time to last unformed stool and diarrheal frequency) conditions in children, adolescents and adults. A. clausii 088AE treatment decreased AAD-severity score on visit 5 in both pediatric (0.12 ± 0.33, 12.39 folds), adult and adolescent (0.54 ± 0.36, 2.34 folds) groups compared to those respective placebo arm (p < 0.05). A. clausii 088AE was well tolerated, did not cause significant changes in vital and clinical safety parameters and subjects reported no adverse effects or serious adverse reactions. A. clausii 088AE is safe and therapeutically effective against AAD, reducing onset of diarrhea and related severity symptoms including abdominal discomfort and pain, bloating and flatulence. A. clausii 088AE may be recommended as a live bio-therapeutic agent for improving clinical pathophysiology of gastrointestinal ailments, in particular antibiotic-associated diarrhea and related symptoms.
Collapse
Affiliation(s)
- Chiranjit Maity
- Probiotics Laboratory, Advanced Enzyme Technologies Ltd., 5Th Floor, A-Wing, Sun Magnetica, LIC Service Road, Louiswadi, Thane (W), 400 604, Maharashtra, India
| | - Anil Kumar Gupta
- Probiotics Laboratory, Advanced Enzyme Technologies Ltd., 5Th Floor, A-Wing, Sun Magnetica, LIC Service Road, Louiswadi, Thane (W), 400 604, Maharashtra, India
| |
Collapse
|
231
|
Rajković E, Schwarz C, Tischler D, Schedle K, Reisinger N, Emsenhuber C, Ocelova V, Roth N, Frieten D, Dusel G, Gierus M. Potential of Grape Extract in Comparison with Therapeutic Dosage of Antibiotics in Weaning Piglets: Effects on Performance, Digestibility and Microbial Metabolites of the Ileum and Colon. Animals (Basel) 2021; 11:ani11102771. [PMID: 34679793 PMCID: PMC8532789 DOI: 10.3390/ani11102771] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/03/2021] [Accepted: 09/17/2021] [Indexed: 11/27/2022] Open
Abstract
Simple Summary Diarrhea as a symptom of different enteric infections leads to poor animal health and performance at weaning, followed by economic losses. Phytogenic feed additives, e.g., grape extracts, have shown antimicrobial and anti-inflammatory properties and these might have beneficial effects on growth trends of weaning piglets and, thereby, potentially reduce the need for antibiotic treatments following weaning. An 8-week feeding trial investigated the potential effects of grape extract (GE) in a model with a negative control (NC) and positive control (PC; antibiotic treatment). Despite no changes in animal performance, dietary GE improved the digestibility of selected nutrients at the same, or even at higher level, as PC. Additionally, there was no clear effect of dietary intervention on the microbial metabolites from the ileum and colon at the end of the trial. These results indicated beneficial effects of GE compared to antibiotic treatment, as often applied at weaning. Abstract Enteric diseases in piglets, such as post-weaning diarrhea (PWD), often require antibiotic treatment of the entire litter. Grape polyphenols may help overcome PWD and thereby reduce the need for antibiotics. The potential of a grape extract (GE; continuous in-feed supplementation) on performance of weaning piglets, compared with both negative (NC; corn-based diet) and positive control (PC; NC + in-feed antibiotic (amoxicillin) in a therapeutic dosage for day 1–day 5 post weaning) was assessed. Apparent total tract digestibility (ATTD) and microbial metabolites were also evaluated on two sampling points (day 27/28 and day 55/56). We assigned 180 weaning piglets (6.9 ± 0.1 kg body weight (BW)) to 6 male and 6 female pens per treatment with 5 piglets each. Animals from PC showed higher BW on day 13 compared with NC and GE, and a tendency for higher BW on day 56 (p = 0.080) compared to NC. Furthermore, PC increased the average daily feed intake in the starter phase (day 1–day 13), and the average daily gain in the early grower phase (day 14–day 24). Overall, GE improved the ATTD at the same level as PC (ash, acid-hydrolyzed ether extract), or at a higher level than PC (dry matter, organic matter, gross energy, crude protein, P). There were no effects on microbial metabolites apart from minor trends for lactic acid and ammonia. Dietary inclusion of GE may have beneficial effects compared to therapeutic antibiotics, as frequently used at weaning.
Collapse
Affiliation(s)
- Emina Rajković
- FFoQSI GmbH—Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, 3430 Tulln, Austria; (E.R.); (D.T.)
- Department of Agrobiotechnology, Institute of Animal Nutrition, Livestock Products, and Nutrition Physiology (TTE), IFA-Tulln, University of Natural Resources and Life Sciences, Vienna (BOKU), 1190 Vienna, Austria; (K.S.); (M.G.)
| | - Christiane Schwarz
- Department of Agrobiotechnology, Institute of Animal Nutrition, Livestock Products, and Nutrition Physiology (TTE), IFA-Tulln, University of Natural Resources and Life Sciences, Vienna (BOKU), 1190 Vienna, Austria; (K.S.); (M.G.)
- Correspondence: ; Tel.: +43-1-47654-97615
| | - David Tischler
- FFoQSI GmbH—Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, 3430 Tulln, Austria; (E.R.); (D.T.)
- Department of Agrobiotechnology, Institute of Animal Nutrition, Livestock Products, and Nutrition Physiology (TTE), IFA-Tulln, University of Natural Resources and Life Sciences, Vienna (BOKU), 1190 Vienna, Austria; (K.S.); (M.G.)
| | - Karl Schedle
- Department of Agrobiotechnology, Institute of Animal Nutrition, Livestock Products, and Nutrition Physiology (TTE), IFA-Tulln, University of Natural Resources and Life Sciences, Vienna (BOKU), 1190 Vienna, Austria; (K.S.); (M.G.)
| | | | | | | | - Nataliya Roth
- BIOMIN Holding GmbH, 3131 Getzersdorf, Austria; (V.O.); (N.R.)
| | - Dörte Frieten
- Department of Animal Nutrition, University of Applied Sciences, 55411 Bingen am Rhein, Germany; (D.F.); (G.D.)
| | - Georg Dusel
- Department of Animal Nutrition, University of Applied Sciences, 55411 Bingen am Rhein, Germany; (D.F.); (G.D.)
| | - Martin Gierus
- Department of Agrobiotechnology, Institute of Animal Nutrition, Livestock Products, and Nutrition Physiology (TTE), IFA-Tulln, University of Natural Resources and Life Sciences, Vienna (BOKU), 1190 Vienna, Austria; (K.S.); (M.G.)
| |
Collapse
|
232
|
The Role of Gut Microbiota and Gut-Brain Interplay in Selected Diseases of the Central Nervous System. Int J Mol Sci 2021; 22:ijms221810028. [PMID: 34576191 PMCID: PMC8471822 DOI: 10.3390/ijms221810028] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome has attracted increasing attention from researchers in recent years. The microbiota can have a specific and complex cross-talk with the host, particularly with the central nervous system (CNS), creating the so-called “gut–brain axis”. Communication between the gut, intestinal microbiota, and the brain involves the secretion of various metabolites such as short-chain fatty acids (SCFAs), structural components of bacteria, and signaling molecules. Moreover, an imbalance in the gut microbiota composition modulates the immune system and function of tissue barriers such as the blood–brain barrier (BBB). Therefore, the aim of this literature review is to describe how the gut–brain interplay may contribute to the development of various neurological disorders, combining the fields of gastroenterology and neuroscience. We present recent findings concerning the effect of the altered microbiota on neurodegeneration and neuroinflammation, including Alzheimer’s and Parkinson’s diseases, as well as multiple sclerosis. Moreover, the impact of the pathological shift in the microbiome on selected neuropsychological disorders, i.e., major depressive disorders (MDD) and autism spectrum disorder (ASD), is also discussed. Future research on the effect of balanced gut microbiota composition on the gut–brain axis would help to identify new potential opportunities for therapeutic interventions in the presented diseases.
Collapse
|
233
|
Kabwe M, Dashper S, Bachrach G, Tucci J. Bacteriophage manipulation of the microbiome associated with tumour microenvironments-can this improve cancer therapeutic response? FEMS Microbiol Rev 2021; 45:6188389. [PMID: 33765142 DOI: 10.1093/femsre/fuab017] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Some cancer treatment failures have been attributed to the tumour microbiota, with implications that microbiota manipulation may improve treatment efficacy. While antibiotics have been used to control bacterial growth, their dysbiotic effects on the microbiome, failure to penetrate biofilms and decreased efficacy due to increasing antimicrobial resistance by bacteria, suggest alternatives are needed. Bacteriophages may provide a precise means for targeting oncobacteria whose relative abundance is increased in tumour tissue microbiomes. Fusobacterium, Streptococcus, Peptostreptococcus, Prevotella, Parvimonas, and Treponema species are prevalent in tumour tissue microbiomes of some cancers. They may promote cancer growth by dampening immunity, stimulating release of proinflammatory cytokines, and directly interacting with cancer cells to stimulate proliferation. Lytic bacteriophages against some of these oncobacteria have been isolated and characterised. The search continues for others. The possibility exists for their testing as adjuncts to complement existing therapies. In this review, we highlight the role of oncobacteria, specifically those whose relative abundance in the intra-tumour microbiome is increased, and discuss the potential for bacteriophages against these micro-organisms to augment existing cancer therapies. The capacity for bacteriophages to modulate immunity and kill specific bacteria makes them suitable candidates to manipulate the tumour microbiome and negate the effects of these oncobacteria.
Collapse
Affiliation(s)
- Mwila Kabwe
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Sharon St. Bendigo, Victoria 3550, Australia
| | - Stuart Dashper
- Melbourne Dental School, University of Melbourne, 720 Swanston St, Parkville, Victoria 3010, Australia
| | - Gilad Bachrach
- The Institute of Dental Sciences, The Hebrew University-Hadassah School of Dental Medicine, PO Box 12272, Jerusalem 9112102, Israel
| | - Joseph Tucci
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Sharon St. Bendigo, Victoria 3550, Australia
| |
Collapse
|
234
|
Huo D, Cen C, Chang H, Ou Q, Jiang S, Pan Y, Chen K, Zhang J. Probiotic Bifidobacterium longum supplied with methimazole improved the thyroid function of Graves' disease patients through the gut-thyroid axis. Commun Biol 2021; 4:1046. [PMID: 34493790 PMCID: PMC8423791 DOI: 10.1038/s42003-021-02587-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
Abstract
Graves’ disease (GD) is an autoimmune disorder that frequently results in hyperthyroidism and other symptoms. Here, we designed a 6-month study with patients divided into three treatment groups, namely, methimazole (MI, n = 8), MI + black bean (n = 9) and MI + probiotic Bifidobacterium longum (n = 9), to evaluate the curative effects of probiotics supplied with MI on thyroid function of patients with GD through clinical index determination and intestinal microbiota metagenomic sequencing. Unsurprisingly, MI intake significantly improved several thyroid indexes but not the most important thyrotropin receptor antibody (TRAb), which is an indicator of the GD recurrence rate. Furthermore, we observed a dramatic response of indigenous microbiota to MI intake, which was reflected in the ecological and evolutionary scale of the intestinal microbiota. In contrast, we did not observe any significant changes in the microbiome in the MI + black bean group. Similarly, the clinical thyroid indexes of patients with GD in the probiotic supplied with MI treatment group continued to improve. Dramatically, the concentration of TRAb recovered to the healthy level. Further mechanistic exploration implied that the consumed probiotic regulated the intestinal microbiota and metabolites. These metabolites impacted neurotransmitter and blood trace elements through the gut-brain axis and gut-thyroid axis, which finally improved the host’s thyroid function. Huo et al conducted a 6 month study in patients with Graves’ Disease in which they administered probiotic Bifidobacterium longum with and without methimazole. They report that Bifidobacterium longum with methimazole potentially improves thyroid function in patients and provide evidence to suggest that this is via the gut-thyroid axis.
Collapse
Affiliation(s)
- Dongxue Huo
- Department of Endocrinology, Hainan General Hospital, School of Food Science and Engineering, Hainan University, Haikou, China.,Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, China
| | - Chaoping Cen
- Department of Endocrinology, Hainan General Hospital, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Haibo Chang
- Department of Endocrinology, Hainan General Hospital, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Qianying Ou
- Department of Endocrinology, Hainan General Hospital, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Shuaiming Jiang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, China
| | - Yonggui Pan
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, China
| | - Kaining Chen
- Department of Endocrinology, Hainan General Hospital, School of Food Science and Engineering, Hainan University, Haikou, China.
| | - Jiachao Zhang
- Department of Endocrinology, Hainan General Hospital, School of Food Science and Engineering, Hainan University, Haikou, China. .,Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, China.
| |
Collapse
|
235
|
Effect of Intrapartum Antibiotics Prophylaxis on the Bifidobacterial Establishment within the Neonatal Gut. Microorganisms 2021; 9:microorganisms9091867. [PMID: 34576761 PMCID: PMC8471514 DOI: 10.3390/microorganisms9091867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022] Open
Abstract
Antibiotics are important disruptors of the intestinal microbiota establishment, linked to immune and metabolic alterations. The intrapartum antibiotics prophylaxis (IAP) is a common clinical practice that is present in more than 30% of labours, and is known to negatively affect the gut microbiota composition. However, little is known about how it affects to Bifidobacterium (sub)species level, which is one of the most important intestinal microbial genera early in life. This study presents qualitative and quantitative analyses of the bifidobacterial (sub)species populations in faecal samples, collected at 2, 10, 30 and 90 days of life, from 43 healthy full-term babies, sixteen of them delivered after IAP use. This study uses both 16S rRNA–23S rRNA internal transcribed spacer (ITS) region sequencing and q-PCR techniques for the analyses of the relative proportions and absolute levels, respectively, of the bifidobacterial populations. Our results show that the bifidobacterial populations establishment is affected by the IAP at both quantitative and qualitative levels. This practice can promote higher bifidobacterial diversity and several changes at a compositional level. This study underlines specific targets for developing gut microbiota-based products for favouring a proper bifidobacterial microbiota development when IAP is required.
Collapse
|
236
|
El-Sayed A, Aleya L, Kamel M. Microbiota and epigenetics: promising therapeutic approaches? ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:49343-49361. [PMID: 34319520 PMCID: PMC8316543 DOI: 10.1007/s11356-021-15623-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/20/2021] [Indexed: 04/15/2023]
Abstract
The direct/indirect responsibility of the gut microbiome in disease induction in and outside the digestive tract is well studied. These results are usually from the overpopulation of certain species on the cost of others, interaction with beneficial microflora, interference with normal epigenetic control mechanisms, or suppression of the immune system. Consequently, it is theoretically possible to cure such disorders by rebalancing the microbiome inside our bodies. This can be achieved by changing the lifestyle pattern and diet or by supplementation with beneficial bacteria or their metabolites. Various approaches have been explored to manipulate the normal microbial inhabitants, including nutraceutical, supplementations with prebiotics, probiotics, postbiotics, synbiotics, and antibiotics, or through microbiome transplantation (fecal, skin, or vaginal microbiome transplantation). In the present review, the interaction between the microbiome and epigenetics and their role in disease induction is discussed. Possible future therapeutic approaches via the reestablishment of equilibrium in our internal micro-ecosystem are also highlighted.
Collapse
Affiliation(s)
- Amr El-Sayed
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030, Besançon Cedex, France
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| |
Collapse
|
237
|
Lu SSM, Mohammed Z, Häggström C, Myte R, Lindquist E, Gylfe Å, Van Guelpen B, Harlid S. Antibiotics Use and Subsequent Risk of Colorectal Cancer: A Swedish Nationwide Population-Based Study. J Natl Cancer Inst 2021; 114:38-46. [PMID: 34467395 PMCID: PMC8755503 DOI: 10.1093/jnci/djab125] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/12/2021] [Accepted: 06/21/2021] [Indexed: 01/12/2023] Open
Abstract
Background Antibiotics use may increase colorectal cancer (CRC) risk by altering the gut microbiota, with suggestive evidence reported. Our study aims to investigate antibiotics use in relation to subsequent CRC risk. Methods This is a nationwide, population-based study with a matched case-control design (first primary CRC cases and 5 matched, cancer-free controls). Complete-population data, extracted from Swedish national registers for the period 2005-2016, were used to calculate odds ratios and 95% confidence intervals. Results We included 40 545 CRC cases and 202 720 controls. Using the full dataset, we found a positive association between more frequent antibiotics use and CRC, excluding antibiotics prescribed within 2 years of diagnosis attenuated results toward the null. In site-specific analyses, excluding the 2-year washout, the positive association was confined to the proximal colon (adjusted odds ratio for very high use vs no use = 1.17, 95% confidence interval = 1.05 to 1.31). For rectal cancer, an inverse association, which appears to be driven by women, was observed. Quinolones and sulfonamides and/or trimethoprims were positively associated with proximal colon cancer, whereas a more general inverse association, across antibiotics classes, was observed for rectal cancer. We found no association between methenamine hippurate, a urinary tract antiseptic not affecting the gut microbiota, and CRC risk. Conclusions This register-based study covering the entire population of Sweden found a robust association between antibiotics use and higher risk of proximal colon cancer and an inverse association with rectal cancer in women. This study strengthens the evidence from previous investigations and adds important insight into site-specific colorectal carcinogenesis.
Collapse
Affiliation(s)
- Sai San Moon Lu
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden.,Department of Epidemiology and Global Health, Umeå University, Umeå, Sweden
| | - Zahraa Mohammed
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Christel Häggström
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Robin Myte
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | | | - Åsa Gylfe
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden.,Umeå Centre for Microbial Research, Umeå University, Umeå, Sweden.,Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden.,Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Sophia Harlid
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| |
Collapse
|
238
|
Gunnlaugsdottir MR, Linnet K, Jonsson JS, Blondal AB. Encouraging rational antibiotic prescribing behaviour in primary care - prescribing practice among children aged 0-4 years 2016-2018: an observational study. Scand J Prim Health Care 2021; 39:373-381. [PMID: 34348560 PMCID: PMC8475099 DOI: 10.1080/02813432.2021.1958506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE To study antibiotic prescriptions among 0- to 4-year-old children before and after implementing a quality project on prudent prescribing of antibiotics in primary healthcare in the capital region of Iceland. DESIGN An observational, descriptive, retrospective study using quantitative methodology. SETTING Primary healthcare in the Reykjavik area with a total population of approximately 220,000. SUBJECTS A total of 6420 children 0-4 years of age presenting at the primary healthcare centres in the metropolitan area over three years from 2016 to 2018. MAIN OUTCOME MEASURES Reduction of antibiotic prescriptions and change in antibiotic profile. Data on antibiotic prescriptions for children 0-4 years of age was obtained from the medical records. Out-of-hours prescriptions were not included in the database. RESULTS The number of prescriptions during the study period ranged from 263.6 to 289.6 prescriptions/1000 inhabitants/year. A reduction of 9% in the total number of prescriptions between 2017-2018 was observed. More than half of all prescriptions were for otitis media, followed by pneumonia and skin infections. Amoxicillin accounted for over half of all prescriptions, increasing between 2016 and 2018 by 51.3%. During this period, the prescribing of co-amoxiclav and macrolides decreased by 52.3% and 40.7%, respectively. These changes were significant in all cases, p < 0.0001. CONCLUSION The results show an overall decrease in antibiotic prescribing concurrent with a change in the choice of antibiotics prescribed and in line with the recommendations presented in the prescribing guidelines implemented by the Primary Healthcare of the Capital Area, and consistent with the project's goals.Key pointsA substantial proportion of antibiotic prescribing can be considered inappropriate and the antibiotic prescription rate is highest in Iceland of the Nordic countries.After implementing guidance on the treatment of common infections together with feedback on antibiotic prescribing, a decrease in the total number of prescriptions accompanied by a shift in the antibiotic profile was observed.
Collapse
Affiliation(s)
| | | | - Jon Steinar Jonsson
- Development Centre for Primary Healthcare, Iceland
- Department of Family Medicine, University of Iceland, Reykjavík, Iceland
| | - Anna Bryndis Blondal
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavík, Iceland
- Development Centre for Primary Healthcare, Iceland
- CONTACT Anna Bryndis Blondal , Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavík, Iceland
| |
Collapse
|
239
|
Martín-Núñez GM, Cornejo-Pareja I, Clemente-Postigo M, Tinahones FJ, Moreno-Indias I. Helicobacter pylori Eradication Therapy Affect the Gut Microbiota and Ghrelin Levels. Front Med (Lausanne) 2021; 8:712908. [PMID: 34458288 PMCID: PMC8387937 DOI: 10.3389/fmed.2021.712908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Antibiotic therapy used to eradicate Helicobacter pylori has been associated with changes in plasma ghrelin and alterations in the gut microbiota. On the other hand, changes in ghrelin levels have been related to changes in gut microbiota composition. Our aim was to evaluate the relationship between changes in the gut microbiota and ghrelin levels in H. pylori infected patients who received antibiotic treatment for its eradication. Methods: A prospective case-control study that included forty H. pylori-positive patients who received eradication therapy (omeprazole, clarithromycin, and amoxicillin) and twenty healthy H. pylori antigen-negative participants. Patients were evaluated, including clinical, anthropometric and dietary variables, before and 2 months after treatment. Gut microbiota composition was analyzed through 16S rRNA amplicon sequencing (IlluminaMiSeq). Results: Changes in gut microbiota profiles and decrease in ghrelin levels were identified after H. pylori eradication treatment. Gut bacteria such as Bifidobacterium longum, Bacteroides, Prevotella, Parabacteroides distasonis, and RS045 have been linked to ghrelin levels fasting and/or post meals. Changes in the abundance of Lachnospiraceae, its genus Blautia, as well as Prevotella stercorea, and Megasphaera have been inversely associated with changes in ghrelin after eradication treatment. Conclusions: Eradication treatment for H. pylori produces changes in the composition of the intestinal microbiota and ghrelin levels. The imbalance between lactate producers such as Blautia, and lactate consumers such as Megasphaera, Lachnospiraceae, or Prevotella, could trigger changes related to ghrelin levels under the alteration of the eradication therapy used for H. pylori. In addition, acetate producing bacteria such as B. longum, Bacteroides, and P. distasonis could also play an important role in ghrelin regulation.
Collapse
Affiliation(s)
- Gracia Mª Martín-Núñez
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Cornejo-Pareja
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes Clemente-Postigo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.,Department of Cell Biology, Physiology, and Immunology, Maimónides Biomedical Research Institute of Córdoba (IMIBIC)/University of Córdoba/Reina Sofia University Hospital, Córdoba, Spain
| | - Francisco J Tinahones
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Isabel Moreno-Indias
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain.,Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
240
|
Induri SNR, Kansara P, Thomas SC, Xu F, Saxena D, Li X. The Gut Microbiome, Metformin, and Aging. Annu Rev Pharmacol Toxicol 2021; 62:85-108. [PMID: 34449247 DOI: 10.1146/annurev-pharmtox-051920-093829] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metformin has been extensively used for the treatment of type 2 diabetes, and it may also promote healthy aging. Despite its widespread use and versatility, metformin's mechanisms of action remain elusive. The gut typically harbors thousands of bacterial species, and as the concentration of metformin is much higher in the gut as compared to plasma, it is plausible that microbiome-drug-host interactions may influence the functions of metformin. Detrimental perturbations in the aging gut microbiome lead to the activation of the innate immune response concomitant with chronic low-grade inflammation. With the effectiveness of metformin in diabetes and antiaging varying among individuals, there is reason to believe that the gut microbiome plays a role in the efficacy of metformin. Metformin has been implicated in the promotion and maintenance of a healthy gut microbiome and reduces many age-related degenerative pathologies. Mechanistic understanding of metformin in the promotion of a healthy gut microbiome and aging will require a systems-level approach. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sri Nitya Reddy Induri
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Payalben Kansara
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Scott C Thomas
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Fangxi Xu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA; .,Department of Surgery, New York University School of Medicine, New York, NY 10016, USA
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA;
| |
Collapse
|
241
|
Patel BK, Patel KH, Bhatia M, Iyer SG, Madhavan K, Moochhala SM. Gut microbiome in acute pancreatitis: A review based on current literature. World J Gastroenterol 2021; 27:5019-5036. [PMID: 34497432 PMCID: PMC8384740 DOI: 10.3748/wjg.v27.i30.5019] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/04/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome is a complex microbial community, recognized for its potential role in physiology, health, and disease. The available evidence supports the role of gut dysbiosis in pancreatic disorders, including acute pancreatitis (AP). In AP, the presence of gut barrier damage resulting in increased mucosal permeability may lead to translocation of intestinal bacteria, necrosis of pancreatic and peripancreatic tissue, and infection, often accompanied by multiple organ dysfunction syndrome. Preserving gut microbial homeostasis may reduce the systemic effects of AP. A growing body of evidence suggests the possible involvement of the gut microbiome in various pancreatic diseases, including AP. This review discusses the possible role of the gut microbiome in AP. It highlights AP treatment and supplementation with prebiotics, synbiotics, and probiotics to maintain gastrointestinal microbial balance and effectively reduce hospitalization, morbidity and mortality in an early phase. It also addresses novel therapeutic areas in the gut microbiome, personalized treatment, and provides a roadmap of human microbial contributions to AP that have potential clinical benefit.
Collapse
Affiliation(s)
- Bharati Kadamb Patel
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore
| | - Kadamb H Patel
- School of Applied Sciences, Temasek Polytechnic, Singapore 529757, Singapore
| | - Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch 8140, New Zealand
| | - Shridhar Ganpati Iyer
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- National University Hospital, National University of Singapore, Singapore 119228, Singapore
| | - Krishnakumar Madhavan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- National University Hospital, National University of Singapore, Singapore 119228, Singapore
| | - Shabbir M Moochhala
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
242
|
Pernomian L, Duarte-Silva M, de Barros Cardoso CR. The Aryl Hydrocarbon Receptor (AHR) as a Potential Target for the Control of Intestinal Inflammation: Insights from an Immune and Bacteria Sensor Receptor. Clin Rev Allergy Immunol 2021; 59:382-390. [PMID: 32279195 DOI: 10.1007/s12016-020-08789-3] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aryl hydrocarbon receptor (AHR) is widely expressed in immune and non-immune cells of the gut and its activation has been correlated to the outcome of inflammatory bowel diseases (IBD). In ulcerative colitis and Crohn's disease, there is an excessive chronic inflammation with massive accumulation of leukocytes in the gut, in an attempt to constrain the invasion of pathogenic microorganisms on the damaged organ. Accordingly, it is known that dietary components, xenobiotics, and some chemicals or metabolites can activate AHR and induce the modulation of inflammatory responses. In fact, the AHR triggering by specific ligands during inflammatory conditions results in decreased IFNγ, IL-6, IL-12, TNF, IL-7, and IL-17, along with reduced microbial translocation and fibrosis in the gut. Moreover, upon AHR activation, there are increased regulatory mechanisms such as IL-10, IL-22, prostaglandin E2, and Foxp3, besides the production of anti-microbial peptides and epithelial repair. Most interestingly, commensal bacteria or their metabolites may also activate this receptor, thus contributing to the restoration of gut normobiosis and homeostasis. In line with that, Lactobacillus reuteri, Lactobacillus bulgaricus, or microbial products such as tryptophan metabolites, indole-3-pyruvic acid, urolithin A, short-chain fatty acids, dihydroxyquinoline, and others may regulate the inflammation by mechanisms dependent on AHR activation. Hence, here we discussed the potential modulatory role of AHR on intestinal inflammation, focused on the reestablishment of homeostasis through the receptor triggering by microbial metabolites. Finally, the development of AHR-based therapies derived from bacteria products could represent an important future alternative for controlling IBD.
Collapse
Affiliation(s)
- Larissa Pernomian
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Murillo Duarte-Silva
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Cristina Ribeiro de Barros Cardoso
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
243
|
Gu X, Yu T, Guo T, Kong J. A qPCR-based method for rapid quantification of six intestinal homeostasis-relevant bacterial genera in feces. Future Microbiol 2021; 16:895-906. [PMID: 34342236 DOI: 10.2217/fmb-2020-0269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Developing efficient methods for monitoring the complex microbial community to rapidly assess the health status. Materials & methods: The qPCR-based method was developed, verified and in situ applied in fecal samples. Results: Six primer pairs with high specificity were designed to perform qPCR assays under a unified reaction condition within 2.5 h. The limits of detection, amplification efficiency and feasibility of the qPCR-based method established here were verified. In situ application of 18 fecal samples showed that the amounts of Bacteroides, Streptococcus and Bifidobacterium in colorectal cancer patient feces were obviously lower than those of healthy volunteers. Conclusion: This qPCR-based method was a reliable tool for rapid quantification of the six intestinal homeostasis relevant bacterial genera in feces.
Collapse
Affiliation(s)
- Xinyi Gu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Tao Yu
- Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Tingting Guo
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Jian Kong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| |
Collapse
|
244
|
Freccero F, Lanci A, Mariella J, Viciani E, Quercia S, Castagnetti A, Castagnetti C. Changes in the Fecal Microbiota Associated with a Broad-Spectrum Antimicrobial Administration in Hospitalized Neonatal Foals with Probiotics Supplementation. Animals (Basel) 2021; 11:ani11082283. [PMID: 34438741 PMCID: PMC8388449 DOI: 10.3390/ani11082283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Post-antibiotic intestinal dysbiosis leads to an overall reduction in bacterial and functional diversity, along with a minor resistance against pathogens. The study aimed to determine the changes on the fecal microbiota in hospitalized neonatal foals administered with broad-spectrum antimicrobials and supplemented probiotics. Fecal samples were collected at hospital admission, at the end of the antimicrobial treatment and at discharge. Seven foals treated with intravenous ampicillin and aminoglycosides for a mean of seven days were included. The results suggest that the fecal microbiota of neonatal foals rapidly returns to a high diversity after treatment. While the findings need to be confirmed in a larger population, the study suggests that in foals, the effect of antimicrobials may be strongly influenced by the changes that occur over time in the developing gut microbiota. Of note, the findings are influenced by the use of probiotics, and whether the changes would be consistent in antimicrobial-administered but not supplemented foals remains to be elucidated. Abstract There is a wide array of evidence across species that exposure to antibiotics is associated with dysbiosis, and due to their widespread use, this also raises concerns also in medicine. The study aimed to determine the changes on the fecal microbiota in hospitalized neonatal foals administered with broad-spectrum antimicrobials and supplemented probiotics. Fecal samples were collected at hospital admission (Ta), at the end of the antimicrobial treatment (Te) and at discharge (Td). Feces were analysed by next-generation sequencing of the 16S rRNA gene on Illumina MiSeq. Seven foals treated with IV ampicillin and amikacin/gentamicin were included. The mean age at Ta was 19 h, the mean treatment length was 7 days and the mean time between Te and Td was 4.3 days. Seven phyla were identified: Actinobacteria, Bacteroidetes, Firmicutes, Fusobacteria, Proteobacteria, TM7 and Verrucomicrobia. At Ta, Firmicutes (48.19%) and Proteobacteria (31.56%) were dominant. The alpha diversity decreased from Ta to Te, but it was the highest at Td. The beta diversity was higher at Ta than at Te and higher at Td than at Te. An increase in Akkermansia over time was detected. The results suggest that the intestinal microbiota of neonatal foals rapidly returns to a high diversity after treatment. It is possible that in foals, the effect of antimicrobials is strongly influenced or overshadowed by the time-dependent changes in the developing gut microbiota.
Collapse
Affiliation(s)
- Francesca Freccero
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
| | - Aliai Lanci
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
- Correspondence:
| | - Jole Mariella
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
| | - Elisa Viciani
- Wellmicro srl, Via Piero Gobetti 101, 40127 Bologna, Italy; (E.V.); (S.Q.); (A.C.)
| | - Sara Quercia
- Wellmicro srl, Via Piero Gobetti 101, 40127 Bologna, Italy; (E.V.); (S.Q.); (A.C.)
| | - Andrea Castagnetti
- Wellmicro srl, Via Piero Gobetti 101, 40127 Bologna, Italy; (E.V.); (S.Q.); (A.C.)
| | - Carolina Castagnetti
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sora 50, Ozzano Dell’Emilia, 40064 Bologna, Italy; (F.F.); (J.M.); (C.C.)
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, Ozzano dell’Emilia, 40064 Bologna, Italy
| |
Collapse
|
245
|
Vetrano DL, Triolo F, Maggi S, Malley R, Jackson TA, Poscia A, Bernabei R, Ferrucci L, Fratiglioni L. Fostering healthy aging: The interdependency of infections, immunity and frailty. Ageing Res Rev 2021; 69:101351. [PMID: 33971332 PMCID: PMC9588151 DOI: 10.1016/j.arr.2021.101351] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 12/20/2022]
Abstract
Untangling the interdependency of infections, immunity and frailty may help to clarify their roles in the maintenance of health in aging individuals, and the recent COVID-19 pandemic has further highlighted such priority. In this scoping review we aimed to systematically collect the evidence on 1) the impact of common infections such as influenza, pneumonia and varicella zoster on frailty development, and 2) the role played by frailty in the response to immunization of older adults. Findings are discussed under a unifying framework to identify knowledge gaps and outline their clinical and public health implications to foster a healthier aging. Twenty-nine studies (113,863 participants) selected to answer the first question provided a moderately strong evidence of an association between infections and physical as well as cognitive decline - two essential dimensions of frailty. Thirteen studies (34,520 participants) investigating the second aim, showed that frailty was associated with an impaired immune response in older ages, likely due to immunosenescence. However, the paucity of studies, the absence of tools to predict vaccine efficacy, and the lack of studies investigating the efficacy of newer vaccines in presence of frailty, strongly limit the formulation of more personalized immunization strategies for older adults. The current evidence suggests that infections and frailty repeatedly cross each other pathophysiological paths and accelerate the aging process in a vicious circle. Such evidence opens to several considerations. First, the prevention of both conditions pass through a life course approach, which includes several individual and societal aspects. Second, the maintenance of a well-functioning immune system may be accomplished by preventing frailty, and vice versa. Third, increasing the adherence to immunization may delay the onset of frailty and maintain the immune system homeostasis, beyond preventing infections.
Collapse
Affiliation(s)
- Davide L Vetrano
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Centro Medicina dell'Invecchiamento, Fondazione Policlinico "A- Gemelli" IRCCS and Catholic University of Rome, Italy.
| | - Federico Triolo
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Stefania Maggi
- National Research Council, Neuroscience Institute, Padua, Italy
| | - Richard Malley
- Division of Infectious Diseases, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas A Jackson
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; Department of Geriatrics, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Roberto Bernabei
- Centro Medicina dell'Invecchiamento, Fondazione Policlinico "A- Gemelli" IRCCS and Catholic University of Rome, Italy
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Laura Fratiglioni
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Stockholm Gerontology Research Center, Stockholm, Sweden
| |
Collapse
|
246
|
Liu R, Sun Y, Wu H, Ni S, Wang J, Li T, Bi Y, Feng X, Zhang C, Sun Y. In-depth investigation of the effective substances of traditional Chinese medicine formula based on the novel concept of co-decoction reaction-using Zuojin decoction as a model sample. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1179:122869. [PMID: 34333214 DOI: 10.1016/j.jchromb.2021.122869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/03/2021] [Accepted: 07/20/2021] [Indexed: 01/03/2023]
Abstract
Zuojin decoction (ZJD) is a classic pair composed of Coptidis Rhizoma and Evodiae Fructus, which is suitable for treating gastrointestinal diseases and tumours, etc. In recent years, scientists have been widely focused on research into the treatment of liver cancer using ZJD; however, the effective substances have not yet been comprehensively elucidated. The difference between the co-decoction and the single decoction of ZJD is revealed in this paper based on the UPLC-QE-Orbitrap-MS, and the chemical components absorbed into the blood and liver of mice have been analyzed simultaneously. In addition, the combination of prototype components absorbed into the liver with liver cancer-related targets has been performed via molecular docking to explore the mechanism of ZJD in treating liver cancer. By comparing the co-decoction and single decoction of ZJD, 44 new components appeared during co-decoction and 76 known chemical compounds have been identified at the same time. It has been confirmed that 35 known components and 11 new components were absorbed into the blood. Furthermore, 20 known components were discovered from the sample of liver tissue. Molecular docking results showed that 3-O-feruloylquinic acid has good conjugation with Bcl-2, Stat3, mTOR, and mmp9. Catechin has the lowest binding energy with CDK6 and β-catenin. The study provides data for the further confirmation of the material basis and mechanism of ZJD in treating liver cancer, and provides a new idea for the researches on the compatibility mechanism of prescriptions of traditional Chinese medicine.
Collapse
Affiliation(s)
- Runhua Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shiting Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaqi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tianyi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yuelin Bi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xin Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Chenning Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Yikun Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
247
|
Alexander JL, Mullish BH. A Guide to the Gut Microbiome and its Relevance to Critical Care. ACTA ACUST UNITED AC 2021; 29:1106-1112. [PMID: 33104419 DOI: 10.12968/bjon.2020.29.19.1106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although it is well-established that particular bacteria may cause gastroenteritis and other infections when present in the gut, it is only recently that scientists have made significant inroads into understanding the huge number of other bacteria and additional microbes that live within the gastrointestinal tract, referred to as the gut microbiome. In particular, it is now recognised that bacteria within the gut microbiome have a wide variety of roles in maintaining different aspects of human health, and that disturbances of these bacteria may potentially cause or contribute to a number of different medical conditions, including particular infections, certain cancers, and chronic conditions, including inflammatory bowel disease. Moreover, there is increasing awareness that these bacteria help determine how the body responds to medication, including antibiotics and chemotherapy. There has been growing interest in different approaches to alter the gut microbiome as a novel approach to medical therapy. This article provides an overview of the importance of the gut microbiome, with a particular focus on critical care.
Collapse
Affiliation(s)
- James L Alexander
- Clinical Lecturer and Honorary Specialty Registrar, Department of Metabolism, Digestion and Reproduction, Imperial College London, and Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London
| | - Benjamin H Mullish
- Clinical Lecturer and Honorary Specialty Registrar, Department of Metabolism, Digestion and Reproduction, Imperial College London, and Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London
| |
Collapse
|
248
|
Zhang X, Han Y, Huang W, Jin M, Gao Z. The influence of the gut microbiota on the bioavailability of oral drugs. Acta Pharm Sin B 2021; 11:1789-1812. [PMID: 34386321 PMCID: PMC8343123 DOI: 10.1016/j.apsb.2020.09.013] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/27/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Due to its safety, convenience, low cost and good compliance, oral administration attracts lots of attention. However, the efficacy of many oral drugs is limited to their unsatisfactory bioavailability in the gastrointestinal tract. One of the critical and most overlooked factors is the symbiotic gut microbiota that can modulate the bioavailability of oral drugs by participating in the biotransformation of oral drugs, influencing the drug transport process and altering some gastrointestinal properties. In this review, we summarized the existing research investigating the possible relationship between the gut microbiota and the bioavailability of oral drugs, which may provide great ideas and useful instructions for the design of novel drug delivery systems or the achievement of personalized medicine.
Collapse
Key Words
- 5-ASA, 5-aminosalicylic acid
- AA, ascorbic acid
- ABC, ATP-binding cassette
- ACS, amphipathic chitosan derivative
- AMI, amiodarone
- AQP4, aquaporin 4
- AR, azoreductase
- ASP, amisulpride
- BBR, berberine
- BCRP, breast cancer resistance protein
- BCS, biopharmaceutics classification system
- BDDCS, the biopharmaceutics drug disposition classification system
- BDEPT, the bacteria-directed enzyme prodrug therapy
- BSH, bile salt hydrolase
- Bioavailability
- CA, cholic acid
- CDCA, chenodeoxycholic acid
- CPP, cell-penetrating peptide
- CS, chitosan
- Colon-specific drug delivery system
- DCA, deoxycholic acid
- DRPs, digoxin reduction products
- EcN, Escherichia coli Nissle 1917
- FA, folate
- FAO, Food and Agriculture Organization of the United Nations
- GCDC, glycochenodeoxycholate
- GL, glycyrrhizic acid
- Gut microbiota
- HFD, high fat diet
- HTC, hematocrit
- IBD, inflammatory bowel disease
- LCA, lithocholic acid
- LPS, lipopolysaccharide
- MATEs, multidrug and toxin extrusion proteins
- MDR1, multidrug resistance gene 1
- MDR1a, multidrug resistance protein-1a
- MKC, monoketocholic acid
- MPA, mycophenolic acid
- MRP2, multidrug resistance-associated protein 2
- NEC, necrotizing enterocolitis
- NMEs, new molecular entities
- NRs, nitroreductases
- NSAIDs, non-steroidal anti-inflammatory drugs
- NaDC, sodium deoxycholate
- NaGC, sodium glycholate
- OATs, organic anion transporters
- OCTNs, organic zwitterion/cation
- OCTs, organic cation transporters
- Oral drugs
- P-gp, P-glycoprotein
- PD, Parkinson's disease
- PPIs, proton pump inhibitors
- PT, pectin
- PWSDs, poorly water-soluble drugs
- Probiotics
- RA, rheumatoid arthritis
- RBC, red blood cell
- SCFAs, short-chain fatty acids
- SGLT-1, sodium-coupled glucose transporter 1
- SLC, solute carrier
- SLN, solid lipid nanoparticle
- SP, sulfapyridine
- SSZ, sulfasalazine
- SVCT-1/2, the sodium-dependent vitamin C transporter-1/2
- T1D, type 1 diabetes
- T1DM, type 1 diabetes mellitus
- T2D, type 2 diabetes
- TCA, taurocholate
- TCDC, taurochenodeoxycholate
- TDCA, taurodeoxycholate
- TLCA, taurolithocholate
- TME, the tumor microenvironment
- UDC, ursodeoxycholic acid
- WHO, World Health Organization
- an OTC drug, an over-the-counter drug
- cgr operon, cardiac glycoside reductase operon
- dhBBR, dihydroberberine
- pKa, dissociation constant
- the GI tract, the gastrointestinal tract
Collapse
Affiliation(s)
- Xintong Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
249
|
Padayachee Y, Flicker S, Linton S, Cafferkey J, Kon OM, Johnston SL, Ellis AK, Desrosiers M, Turner P, Valenta R, Scadding GK. Review: The Nose as a Route for Therapy. Part 2 Immunotherapy. FRONTIERS IN ALLERGY 2021; 2:668781. [PMID: 35387044 PMCID: PMC8974912 DOI: 10.3389/falgy.2021.668781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
The nose provides a route of access to the body for inhalants and fluids. Unsurprisingly it has a strong immune defense system, with involvement of innate (e.g., epithelial barrier, muco- ciliary clearance, nasal secretions with interferons, lysozyme, nitric oxide) and acquired (e.g., secreted immunoglobulins, lymphocytes) arms. The lattice network of dendritic cells surrounding the nostrils allows rapid uptake and sampling of molecules able to negotiate the epithelial barrier. Despite this many respiratory infections, including SARS-CoV2, are initiated through nasal mucosal contact, and the nasal mucosa is a significant "reservoir" for microbes including Streptococcus pneumoniae, Neisseria meningitidis and SARS -CoV-2. This review includes consideration of the augmentation of immune defense by the nasal application of interferons, then the reduction of unnecessary inflammation and infection by alteration of the nasal microbiome. The nasal mucosa and associated lymphoid tissue (nasopharynx-associated lymphoid tissue, NALT) provides an important site for vaccine delivery, with cold-adapted live influenza strains (LAIV), which replicate intranasally, resulting in an immune response without significant clinical symptoms, being the most successful thus far. Finally, the clever intranasal application of antibodies bispecific for allergens and Intercellular Adhesion Molecule 1 (ICAM-1) as a topical treatment for allergic and RV-induced rhinitis is explained.
Collapse
Affiliation(s)
- Yorissa Padayachee
- Department of Respiratory Medicine, Faculty of Medicine, Imperial College Healthcare NHS Trust, Imperial College London, London, United Kingdom
| | - Sabine Flicker
- Center for Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Sophia Linton
- Division of Allergy and Immunology, Department of Medicine, Queen's University, Kingston, ON, Canada
- Allergy Research Unit, Kingston Health Sciences Centre (KHSC), Kingston, ON, Canada
| | - John Cafferkey
- Department of Respiratory Medicine, Faculty of Medicine, Imperial College Healthcare NHS Trust, Imperial College London, London, United Kingdom
| | - Onn Min Kon
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sebastian L. Johnston
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Anne K. Ellis
- Division of Allergy and Immunology, Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Martin Desrosiers
- Department of Otorhinolaryngologie, The University of Montreal Hospital Research Centre (CRCHUM), Montreal, QC, Canada
| | - Paul Turner
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Rudolf Valenta
- Division of Immunopathology, Medical University of Vienna, Vienna, Austria
| | - Glenis Kathleen Scadding
- Royal National Ear Nose and Throat Hospital, University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Division of Infection and Immunity, Faculty of Medical Sciences, University College London, London, United Kingdom
| |
Collapse
|
250
|
Wang Z, Fan L, Wang J, Xie S, Zhang C, Zhou J, Zhang L, Xu G, Zou J. Insight into the immune and microbial response of the white-leg shrimp Litopenaeus vannamei to microplastics. MARINE ENVIRONMENTAL RESEARCH 2021; 169:105377. [PMID: 34087762 DOI: 10.1016/j.marenvres.2021.105377] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/07/2021] [Accepted: 05/21/2021] [Indexed: 06/12/2023]
Abstract
Microplastics (MPs) are a new type of environmental pollutant. To investigate the response of shrimp and their microflora to MPs, Litopenaeus vannamei (L. vannamei) was exposed to different concentrations of MPs (0, 50, 500, and 5000 μg/L, i.e., C, L, M and H groups) for 48 h. The survival rate, intake of MPs, immune-related gene expression and microbial response under MP exposure were detected. The results showed that the survival rate in the H group was significantly lower than those in the C, L and M groups, while the relative expression levels of proPO, TLR and ALF in the M and H groups were significantly higher than those in the C and L groups. For the microbial response, microbial community richness in the L group was significantly decreased, while community richness and diversity in the H group were significantly increased compared with those in the C group. The relative abundances of 3, 4 and 11 taxa were significantly changed after MP treatment at the phylum, class and genus levels, respectively. The results suggested that short-term exposure to low concentrations of MPs did not cause immune defense responses or death but affected the balance of bacterial composition in shrimp. Exposure to high concentrations of MPs can induce immune responses and microbial changes and can even cause death in shrimp. These findings increase our understanding of MP impacts on aquatic organisms.
Collapse
Affiliation(s)
- Zhenlu Wang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Lanfen Fan
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Jun Wang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| | - Shaolin Xie
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Chaonan Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Jiang Zhou
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Li Zhang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Guohuan Xu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
| | - Jixing Zou
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|