1
|
Attiq A. Early-life antibiotic exposures: Paving the pathway for dysbiosis-induced disorders. Eur J Pharmacol 2025; 991:177298. [PMID: 39864578 DOI: 10.1016/j.ejphar.2025.177298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/06/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Microbiota encompasses a diverse array of microorganisms inhabiting specific ecological niches. Gut microbiota significantly influences physiological processes, including gastrointestinal motor function, neuroendocrine signalling, and immune regulation. They play a crucial role in modulating the central nervous system and bolstering body defence mechanisms by influencing the proliferation and differentiation of innate and adaptive immune cells. Given the potential consequences of antibiotic therapy on gut microbiota equilibrium, there is a need for prudent antibiotic use to mitigate associated risks. Observational studies have linked increased antibiotic usage to various pathogenic conditions, including obesity, inflammatory bowel disease, anxiety-like effects, asthma, and pulmonary carcinogenesis. Addressing dysbiosis incidence requires proactive measures, including prophylactic use of β-lactamase drugs (SYN-004, SYN-006, and SYN-007), hydrolysing the β-lactam in the proximal GIT for maintaining intestinal flora homeostasis. Prebiotic and probiotic supplementations are crucial in restoring intestinal flora equilibrium by competing with pathogenic bacteria for nutritional resources and adhesion sites, reducing luminal pH, neutralising toxins, and producing antimicrobial agents. Faecal microbiota transplantation (FMT) shows promise in restoring gut microbiota composition. Rational antibiotic use is essential to preserve microflora and improve patient compliance with antibiotic regimens by mitigating associated side effects. Given the significant implications on gut microbiota composition, concerted intervention strategies must be pursued to rectify and reverse the occurrence of antibiotic-induced dysbiosis. Here, antibiotics-induced microbiota dysbiosis mechanisms and their systemic implications are reviewed. Moreover, proposed interventions to mitigate the impact on gut microflora are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| |
Collapse
|
2
|
Li Y, Zhang Y, Zhao J, Zhang X, Liu S, Qi H, Qiao F, Yao H. Isolation and evaluation of Pediococcus acidilactici YH-15 from cat milk: Potential probiotic effects and antimicrobial properties. Heliyon 2024; 10:e39539. [PMID: 39498095 PMCID: PMC11533615 DOI: 10.1016/j.heliyon.2024.e39539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/07/2024] Open
Abstract
The study aimed to screen for the possible presence of lactic acid bacteria (LAB) in cat milk in order to evaluate their probiotic properties. The isolates were characterized by biochemical identification, morphological tests and 16S rDNA sequencing. Afterward, gastrointestinal passage, in vitro safety and probiotic properties were evaluated. The results showed that the isolates had 10 strains of Pediococcus acidilactici permitted in the feed additive catalog. The high survival rate in the acid and bile salt resistance test reflected the good strain tolerance of the isolates to the simulated gastrointestinal conditions of the host in vitro. The mean inhibitory diameters of the 10 strains against chloramphenicol and tetracycline were 23.6 mm and 17.4 mm, respectively; none of the hemolytic tests showed α/β hemolytic ring. The bacteriostatic test showed that P. acidilactici YH-9, YH-14 and YH-15 had inhibitory effects on four common pathogenic bacteria, including Escherichia coli, Salmonella typhimurium, Staphylococcus aureus and Streptococcus. The adhesion test showed that P. acidilactici YH-15 had good adhesion to HT-29 cells. Based on these results, we concluded that P. acidilactici YH-15 extracted from cat milk has potential application as a clinical probiotic therapy and health care product.
Collapse
Affiliation(s)
- Yahui Li
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Yiwen Zhang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Junxin Zhao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Xuan Zhang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Shiwei Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Hanmeng Qi
- Chinese Academy of Sciences Beijing Institute for Stem Cell and Regenerative Medicine, China
| | - Fuqiang Qiao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Hua Yao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| |
Collapse
|
3
|
Zhang Y, Lu B, Dong Y, Zhang Y, Du Q, Chen Y, Zhang Z. Saccharomyces boulardii combined with triple therapy alter the microbiota in the eradication of Helicobacter pylori infection. Sci Rep 2024; 14:13152. [PMID: 38849408 PMCID: PMC11161611 DOI: 10.1038/s41598-024-63894-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
To assess the effectiveness and safety of combining Saccharomyces boulardii powder with triple therapy as a primary approach for eradicating H. pylori infection, a total of 144 patients who tested positive for H. pylori and diagnosed with non-ulcer dyspepsia underwent endoscopy at two national centers between June 2017 and March 2019 were included. The patients were categorized into three groups using a subsection randomization method and received initial H. pylori eradication treatments. Microbial composition, eradication rates, symptom alleviation, and adverse reactions were monitored on the 14th and 44th days post-treatment. According to PP analysis showed the eradication rates for the SRAC group was 75%, BRAC was 93.18% and RAC was 65.2%. Group BRAC exhibited a marginally higher eradication rate compared to other groups. However, patients receiving Saccharomyces boulardii treatment exhibited an overall reduction in initial dyspepsia symptoms by the end of the treatment period. When employed as a primary strategy, the combination of Saccharomyces boulardii powder with triple therapy displayed notable efficacy and smaller gastrointestinal side effects in eradicating initial H. pylori infections among non-ulcer dyspepsia patients. Moreover, this approach demonstrated advantages in alleviating symptoms, exhibited favorable tolerance, and maintained a high level of clinical safety.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, People's Republic of China
| | - Bingyun Lu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, People's Republic of China
| | - Yu Dong
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, People's Republic of China
| | - Yifeng Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, People's Republic of China
| | - Qianming Du
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, People's Republic of China
| | - Ye Chen
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, People's Republic of China.
- Integrative Microecology Clinical Center, Shenzhen Key Laboratory of Gastrointestinal Microbiota and Disease, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Technology Research Center of Gut Microbiota Transplantation, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Bao'an District, Shenzhen, 518101, People's Republic of China.
| | - Zhenyu Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, People's Republic of China.
| |
Collapse
|
4
|
Niu H, Zhou M, Zogona D, Xing Z, Wu T, Chen R, Cui D, Liang F, Xu X. Akkermansia muciniphila: a potential candidate for ameliorating metabolic diseases. Front Immunol 2024; 15:1370658. [PMID: 38571945 PMCID: PMC10987721 DOI: 10.3389/fimmu.2024.1370658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
Metabolic diseases are comprehensive disease based on obesity. Numerous cumulative studies have shown a certain correlation between the fluctuating abundance of Akkermansia muciniphila and the occurrence of metabolic diseases. A. muciniphila, a potential probiotic candidate colonized in the human intestinal mucus layer, and its derivatives have various physiological functions, including treating metabolic disorders and maintaining human health. This review systematically explicates the abundance change rules of A. muciniphila in metabolic diseases. It also details the high efficacy and specific molecules mechanism of A. muciniphila and its derivatives in treating obesity, type 2 diabetes mellitus, cardiovascular disease, and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Huifang Niu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Minfeng Zhou
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Daniel Zogona
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zheng Xing
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ting Wu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Rui Chen
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dandan Cui
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fengxia Liang
- School of Acupuncture and Bone Injury, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaoyun Xu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), Hubei Key Laboratory of Fruit Vegetable Processing Quality Control (Huazhong Agricultural University), School of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
5
|
Paduchová Z, Nagyová Z, Wang D, Muchová J. The impact of probiotics and vitamin C on the prevention of upper respiratory tract symptoms in two preschool children cohorts. Nutr Res Pract 2024; 18:98-109. [PMID: 38352209 PMCID: PMC10861338 DOI: 10.4162/nrp.2024.18.1.98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/16/2023] [Accepted: 11/16/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES The efficacy of Lab4 probiotic and vitamin C combination on the prevention of upper respiratory tract infections (URTIs) was investigated in two studies with children. Our objective was to pool dataset of 57 preschool children from the PROCHILD study (ISRCTN28722693) and the dataset of 50 preschool matched cohort from the PROCHILD-2 study (ISRCTN26587549) to evaluate the impact of probiotic/vitamin C combination on the prevention of upper respiratory tract symptoms and provide a more robust assessment of effect using detailed individual level data. SUBJECTS/METHODS The children were supplemented daily for 6 months with either the multistrain probiotic (1.25×1010 cfu/tablet consisting of two strains of Lactobacillus acidophilus CUL21 and CUL60, Bifidobacterium bifidum CUL20 and Bifidobacterium animalis subsp. lactis CUL34) plus 50 mg vitamin C or a placebo. RESULTS In the pooled analysis of the individual participant data (per protocol population), significant reductions were observed for the incidence (-25%; 95% confidence interval [CI], 0.66, 0.85; P < 0.0001) and duration (-14.9 days; 95% CI, -24.8, -5.1; P = 0.0030) of typical URTI symptoms in the active group compared with the placebo. The incidence rates of absenteeism from preschool (IR ratio, 0.75; 95% CI, 0.66, 0.86; P < 0.0001), paediatric visits (IR ratio, 0.56; 95% CI, 0.47; 0.68; P < 0.0001) and antibiotic usage (IR ratio, 0.53; 95% CI, 0.39, 0.71; P < 0.0001) were also significantly reduced. CONCLUSION The pooled analysis findings of comparable preschool cohorts from two studies indicate that the supplementation with probiotic and vitamin C combination is beneficial in the prevention and management of URTI symptoms.
Collapse
Affiliation(s)
- Zuzana Paduchová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia
| | - Zuzana Nagyová
- JuvenaliaA Paediatric Centre, 929 01 Dunajská Streda, Slovakia
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, United Kingdom
| | - Jana Muchová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, 813 72 Bratislava, Slovakia
| |
Collapse
|
6
|
Paraskevaidis I, Xanthopoulos A, Tsougos E, Triposkiadis F. Human Gut Microbiota in Heart Failure: Trying to Unmask an Emerging Organ. Biomedicines 2023; 11:2574. [PMID: 37761015 PMCID: PMC10526035 DOI: 10.3390/biomedicines11092574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
There is a bidirectional relationship between the heart and the gut. The gut microbiota, the community of gut micro-organisms themselves, is an excellent gut-homeostasis keeper since it controls the growth of potentially harmful bacteria and protects the microbiota environment. There is evidence suggesting that a diet rich in fatty acids can be metabolized and converted by gut microbiota and hepatic enzymes to trimethyl-amine N-oxide (TMAO), a product that is associated with atherogenesis, platelet dysfunction, thrombotic events, coronary artery disease, stroke, heart failure (HF), and, ultimately, death. HF, by inducing gut ischemia, congestion, and, consequently, gut barrier dysfunction, promotes the intestinal leaking of micro-organisms and their products, facilitating their entrance into circulation and thus stimulating a low-grade inflammation associated with an immune response. Drugs used for HF may alter the gut microbiota, and, conversely, gut microbiota may modify the pharmacokinetic properties of the drugs. The modification of lifestyle based mainly on exercise and a Mediterranean diet, along with the use of pre- or probiotics, may be beneficial for the gut microbiota environment. The potential role of gut microbiota in HF development and progression is the subject of this review.
Collapse
Affiliation(s)
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| | - Elias Tsougos
- 6th Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece
| | - Filippos Triposkiadis
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece; (A.X.); (F.T.)
| |
Collapse
|
7
|
Éliás AJ, Barna V, Patoni C, Demeter D, Veres DS, Bunduc S, Erőss B, Hegyi P, Földvári-Nagy L, Lenti K. Probiotic supplementation during antibiotic treatment is unjustified in maintaining the gut microbiome diversity: a systematic review and meta-analysis. BMC Med 2023; 21:262. [PMID: 37468916 DOI: 10.1186/s12916-023-02961-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Probiotics are often used to prevent antibiotic-induced low-diversity dysbiosis, however their effect is not yet sufficiently summarized in this regard. We aimed to investigate the effects of concurrent probiotic supplementation on gut microbiome composition during antibiotic therapy. METHODS We performed a systematic review and meta-analysis of randomized controlled trials reporting the differences in gut microbiome diversity between patients on antibiotic therapy with and without concomitant probiotic supplementation. The systematic search was performed in three databases (MEDLINE (via PubMed), Embase, and Cochrane Central Register of Controlled Trials (CENTRAL)) without filters on 15 October 2021. A random-effects model was used to estimate pooled mean differences (MD) with 95% confidence intervals (CI). This review was registered on PROSPERO (CRD42021282983). RESULTS Of 11,769 identified articles, 15 were eligible in the systematic review and 5 in the meta-analyses. Quantitative data synthesis for Shannon (MD = 0.23, 95% CI: [(-)0.06-0.51]), Chao1 (MD = 11.59 [(-)18.42-41.60]) and observed OTUs (operational taxonomic unit) (MD = 17.15 [(-)9.43-43.73]) diversity indices revealed no significant difference between probiotic supplemented and control groups. Lacking data prevented meta-analyzing other diversity indices; however, most of the included studies reported no difference in the other reported α- and ß-diversity indices between the groups. Changes in the taxonomic composition varied across the eligible studies but tended to be similar in both groups. However, they showed a potential tendency to restore baseline levels in both groups after 3-8 weeks. This is the first meta-analysis and the most comprehensive review of the topic to date using high quality methods. The limited number of studies and low sample sizes are the main limitations of our study. Moreover, there was high variability across the studies regarding the indication of antibiotic therapy and the type, dose, and duration of antimicrobials and probiotics. CONCLUSIONS Our results showed that probiotic supplementation during antibiotic therapy was not found to be influential on gut microbiome diversity indices. Defining appropriate microbiome diversity indices, their standard ranges, and their clinical relevance would be crucial.
Collapse
Affiliation(s)
- Anna Júlia Éliás
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Doctoral School of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Viktória Barna
- Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Cristina Patoni
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Dóra Demeter
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Military Hospital Medical Centre, Hungarian Defense Forces, Budapest, Hungary
| | - Dániel Sándor Veres
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Stefania Bunduc
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Fundeni Clinical Institute, Bucharest, Romania
| | - Bálint Erőss
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - László Földvári-Nagy
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary.
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary.
| | - Katalin Lenti
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| |
Collapse
|
8
|
Kim J, Lee S. Maternal low-intensity exercise and probiotic ingestion during pregnancy improve physical ability and brain function in offspring mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
9
|
Du L, Chen B, Cheng F, Kim J, Kim JJ. Effects of Helicobacter pylori Therapy on Gut Microbiota: A Systematic Review and Meta-Analysis. Dig Dis 2022; 42:102-112. [PMID: 36228588 DOI: 10.1159/000527047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 09/04/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Although indications for evaluation and treatment of Helicobacter pylori infection are broadening to include primary prevention for gastric adenocarcinoma, potential adverse effects on gut microbiota have been raised. We performed a systematic review and meta-analysis to evaluate the effects of H. pylori therapy on gut microbiota. METHODS PubMed, EMBASE, Cochrane Library, and Web of Science (to 4/2021) were searched for studies quantitatively evaluating microbiota before and after H. pylori therapy. Meta-analysis was performed to assess early (<1 year) and long-term (≥1 year) effects on gut microbiota after H. pylori treatment. Subgroup analysis evaluating the effects of H. pylori therapy with addition of probiotics on gut microbiota was also performed. RESULTS Thirty studies (N = 1,218) met the criteria. Early after H. pylori therapy, intestinal microbial diversity was reduced in nearly all studies. At the genus level, reduction in the abundance of Enterococcus, while increase in Lactobacillus, Bifidobacterium, and Bacteroides counts were observed. However, Enterococcus, Lactobacillus, Bifidobacterium, and Bacteroides counts remained stable in patients who received probiotics with H. pylori therapy. At the phylum level, the relative abundance of Actinobacteria and Firmicutes increased after treatment. At ≥1 year, intestinal microbial diversity normalized in six of seven studies. No differences in the relative abundance of Actinobacteria, Firmicute, Bacteroidetes, and Proteobacteria were observed ≥1 year after therapy. CONCLUSION The impact of H. pylori therapy on gut microbiota appears transient with early changes largely resolving after 1 year. Probiotics may reduce the early impact of H. pylori therapy on gut microbiota.
Collapse
Affiliation(s)
- Lijun Du
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China,
| | - Binrui Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fangli Cheng
- Department of Gastroenterology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jeffrey Kim
- Department of Family Medicine, Loma Linda University Health, Loma Linda, California, USA
| | - John J Kim
- Division of Gastroenterology, Loma Linda University Health, Loma Linda, California, USA
| |
Collapse
|
10
|
Chen B, Li XM, Cai T, Wang F. Short-term and long-term alterations of gastrointestinal microbiota with different H. pylori eradication regimens: A meta-analysis. Front Cell Infect Microbiol 2022. [DOI: 10.3389/fcimb.2022.913384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background and AimsThe impacts of Helicobacter pylori (H. pylori) eradication on the gastrointestinal microbiota are controversial, and whether the short-term and long-term changes in the gastrointestinal microbiota following different eradication regimens are consistent remains inconclusive. This study aimed to examine the effects of various eradication regimens on the gastrointestinal microflora at follow-up evaluations within 7 days, at 1–3 months, and over 6 months changes in the gastrointestinal microbiota.Materials and MethodsStudies reported on the PubMed, Embase, Cochrane Library, Web of Science, and ClinicalTrails.gov databases before March 2022 were collected. Data analysis and visualization were conducted using Review Manager 5.4.1. The tool of the Cochrane Collaboration to assess the risk of bias was suitable for randomized controlled trials with the Newcastle–Ottawa scale for nonrandomized controlled trials. In addition, the process was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement.ResultsAfter a series of rigorous screenings, a total of 34 articles with 1,204 participants were included for this review analysis. The results showed changes in the gut microflora at the phylum level or the family and genus levels. After metronidazole-containing triple therapy, the number of Enterobacteriaceae increased at 1–3 months follow-up. After Metronidazole-free triple therapy, Actinobacteria decreased significantly, and this trend lasted for more than 6 months. Within 7 days after eradication treatment, the follow-up results showed a decrease in the number of Lactobacillus. After Bismuth-containing quadruple therapy, the changes in Actinobacteria fluctuated with the follow-up time. The changes in Proteobacteria showed a downward trend lasting for 1–3 months after eradication but returned to baseline levels over 6 months after eradication. Subgroup analyses indicated that host age could influence changes in the gut microbiota.ConclusionDifferent eradication regimens had varied effects on the short-term and long-term abundance of the gastrointestinal microbiota, but the decreasing trend of the microbiota diversity was the same for all regimens at the short-term follow-up. This study summarizes the changes of gut microbiota at different stages after different eradication regimens and hope to provide some references for supplementing probiotics, while further studies is needed to support these findings.Systematic Review Registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42021292726
Collapse
|
11
|
Effect of Treadmill Exercise and Probiotic Ingestion on Motor Coordination and Brain Activity in Adolescent Mice. Healthcare (Basel) 2020; 9:healthcare9010007. [PMID: 33374692 PMCID: PMC7822428 DOI: 10.3390/healthcare9010007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/29/2022] Open
Abstract
High-intensity exercise can lead to chronic fatigue, which reduces athletic performance. On the contrary, probiotic supplements have many health benefits, including improvement of gastrointestinal health and immunoregulation. However, the effects of probiotics combined with exercise interventions on motor functions and brain activity have not been fully explored. Therefore, this study aimed to identify the effects of probiotic supplements and aerobic exercise on motor function, immune response, and exercise intensity and probiotic ingestion. After four weeks of intervention, the motor functions were assessed by rotarod test, then the levels of cytokines, gamma-aminobutyric acid (GABA), and glutamate were detected. The improvement caused by the intake of probiotics in the moderate-intensity exercise group and the non-exercise group in the accelerating mode rotarod was significant (p = 0.038, p < 0.001, respectively). In constant-speed mode, the moderate-intensity exercise group with probiotic ingestion recorded longer runs than the corresponding non-exercise group (p = 0.023), and the improvement owing to probiotics was significant in all groups—non-exercise, moderate, and high-intensity (p = 0.036, p = 0.036, p = 0.012, respectively). The concentrations of inflammatory cytokines were lower, whereas GABA was higher in the probiotics-ingested group. Taken together, exercise and probiotics in adolescence could positively affect brain and motor function.
Collapse
|
12
|
Ye Q, Shao X, Shen R, Chen D, Shen J. Changes in the human gut microbiota composition caused by Helicobacter pylori eradication therapy: A systematic review and meta-analysis. Helicobacter 2020; 25:e12713. [PMID: 32515529 DOI: 10.1111/hel.12713] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/10/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The short-term and long-term effect of Helicobacter pylori (H pylori) eradication on the gut microbiota is controversial; hence, this study aimed to clarify changes in the gut microbiome and microbial diversity after H pylori eradication. MATERIALS AND METHODS Articles published in PubMed, MEDLINE, and EMBASE were searched up to March 20, 2020, with English-language restriction. The outcomes including gut microbiota and alpha diversity were extracted to analysis. And then, Review Manager 5.3 software was used to conduct the data analysis. RESULTS At phylum level, next-generation sequencing was performed. Meta-analysis results showed that Actinobacteria decreased compared with baseline throughout the follow-up period. Proteobacteria increased during short-term follow-up and then returned to normal. In addition, Bacteroidetes decreased and Firmicutes increased only during long-term follow-up. At family or genus level, conventional microbiological culturing was performed. Enterobacteriaceae and Enterococcus both increased during the short-term and interim follow-up. In addition, Lactobacillus only showed a decreasing trend during short-term follow-up, but it appeared statistical decreasing during interim follow-up. Moreover, relatively sufficient evidence showed that alpha diversity decreased during short-term follow-up, and no reliable data were obtained to confirm the change of alpha diversity during interim and long-term follow-up. CONCLUSION In different follow-up periods after H pylori eradication, changes in gut microbiota were inconsistent. Microbial diversity decreased in the short-term follow-up, while there was no data to confirm subsequent alterations. The results provided a basis for the rational selection of probiotics in the eradication process. However, further studies are needed to obtain more clues.
Collapse
Affiliation(s)
- Qunqun Ye
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Xiaona Shao
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Ruiwei Shen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Dawei Chen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Jianwei Shen
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|
13
|
Antibiotic residues and mycotoxins in raw milk in Punjab (India): a rising concern for food safety. Journal of Food Science and Technology 2019; 56:5146-5151. [PMID: 31741539 DOI: 10.1007/s13197-019-03963-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 07/08/2019] [Accepted: 07/16/2019] [Indexed: 12/29/2022]
Abstract
The present study was envisaged with an aim to determine the occurrence of antibiotic residues; enrofloxacin, oxytetracycline, penicillin G, sulphamethoxazole and chloramphenicol as well as mycotoxins; aflatoxin M1 and ochratoxin A in raw milk samples collected from individual animals from dairy farms located in 9 districts of Punjab, India. A total of 168 raw milk samples were collected and analysed using commercially available competitive Enzyme linked immunosorbent assay kits. Out of these, 19 (11.3%) and 9 (5.4%) samples were found positive for antibiotic residues and mycotoxins, respectively. The milk samples were positive for enrofloxacin (4.8%), oxytetracycline (3.0%), penicillin G (3.0%) and sulphamethoxazole (0.6%) residues. The percentage of samples found above maximum residue limit established by Europen Union (EU)/Codex Alimentarius Commission (CAC) were 1.7%, 1.2% and 0.6% for enrofloxacin, oxytetracycline and penicillin G residues, respectively. Aflatoxin M1 was detected in 5.4% of monitored milk samples with 1.2% samples exceeded the established maximum levels of EU but were below the maximum levels established by CAC. The occurrence of antibiotic residues and mycotoxins in raw milk samples above maximum limits is a rising concern for food safety due to possibility of health risks to the consumers.
Collapse
|
14
|
Abstract
Technological developments, including massively parallel DNA sequencing, gnotobiotics, metabolomics, RNA sequencing and culturomics, have markedly propelled the field of microbiome research in recent years. These methodologies can be harnessed to improve our in-depth mechanistic understanding of basic concepts related to consumption of probiotics, including their rules of engagement with the indigenous microbiome and impacts on the human host. We have recently demonstrated that even during probiotic supplementation, resident gut bacteria in a subset of individuals resist the mucosal presence of probiotic strains, limiting their modulatory effect on the microbiome and on the host gut transcriptional landscape. Resistance is partly alleviated by antibiotics treatment, which enables probiotics to interact with the host at the gut mucosal interface, although rather than promoting reconstitution of the indigenous microbiome and of the host transcriptional profile, they inhibit these components from returning to their naïve pre-antibiotic configurations. In this commentary, we discuss our findings in the context of previous and recent works, and suggest that incorporating the state-of-the-art methods currently utilized in microbiome research into the field of probiotics may lead to improved understanding of their mechanisms of activity, as well as their efficacy and long-term safety.
Collapse
Affiliation(s)
- Jotham Suez
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Niv Zmora
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel,Digestive Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel,Internal Medicine Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel,Cancer-Microbiome Research Division, DKFZ, Heidelberg, Germany,CONTACT Eran Elinav Immunology Department, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| |
Collapse
|
15
|
Abdou AM, Hedia RH, Omara ST, Mahmoud MAEF, Kandil MM, Bakry MA. Interspecies comparison of probiotics isolated from different animals. Vet World 2018; 11:227-230. [PMID: 29657408 PMCID: PMC5891879 DOI: 10.14202/vetworld.2018.227-230] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/16/2018] [Indexed: 01/07/2023] Open
Abstract
AIM The aim of the current study was to isolate and identify naturally occurring probiotic Lactobacillus species in different animals with the different environmental background including fish, and farm animals to investigate interspecies differences in probiotics on the species level. MATERIALS AND METHODS A total of 44 fecal and milk samples were collected under aseptic conditions from cattle, buffalo, camel, sheep, goats, and fish. The samples were cultured, and the isolated strains were confirmed biochemically and molecularly using 16S rRNA multiplex polymerase chain reaction (PCR) analysis following DNA extraction from the bacterial isolates. RESULTS A total of 31 isolates identified as lactobacilli were isolated from cattle milk, goat feces, sheep feces, fish feces, buffalo milk, camel milk, and goats' milk. Lactobacillus species were identified based on the size of the PCR product. The results showed that different species were different in their lactobacilli content. At the same time, there were some differences between individuals of the same species. CONCLUSION The diversity of probiotic strains isolated from different animal species implies different types of benefits to the host. Although it would be both money - and time-consuming research, discovering the benefit of each of these strains may provide very important information for the health of both human and animal. Furthermore, transferring these beneficial effects either to individuals within the same species or between different species would be of great importance.
Collapse
Affiliation(s)
- Amr M. Abdou
- Department of Microbiology and Immunology, National Research Centre 12622 Dokki, Giza, Egypt
| | - Riham H. Hedia
- Department of Microbiology and Immunology, National Research Centre 12622 Dokki, Giza, Egypt
| | - Shimaa T. Omara
- Department of Microbiology and Immunology, National Research Centre 12622 Dokki, Giza, Egypt
| | - Mohamed Abd El-Fatah Mahmoud
- Department of Parasitology and Animal Diseases, Division of Veterinary Research, National Research Centre, 12622 Dokki, Giza, Egypt
| | - Mai M. Kandil
- Department of Microbiology and Immunology, National Research Centre 12622 Dokki, Giza, Egypt
| | - M. A. Bakry
- Department of Microbiology and Immunology, National Research Centre 12622 Dokki, Giza, Egypt
| |
Collapse
|
16
|
Goldenberg JZ, Yap C, Lytvyn L, Lo CK, Beardsley J, Mertz D, Johnston BC. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev 2017; 12:CD006095. [PMID: 29257353 PMCID: PMC6486212 DOI: 10.1002/14651858.cd006095.pub4] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Antibiotics can disturb gastrointestinal microbiota which may lead to reduced resistance to pathogens such as Clostridium difficile (C. difficile). Probiotics are live microbial preparations that, when administered in adequate amounts, may confer a health benefit to the host, and are a potential C. difficile prevention strategy. Recent clinical practice guidelines do not recommend probiotic prophylaxis, even though probiotics have the highest quality evidence among cited prophylactic therapies. OBJECTIVES To assess the efficacy and safety of probiotics for preventing C.difficile-associated diarrhea (CDAD) in adults and children. SEARCH METHODS We searched PubMed, EMBASE, CENTRAL, and the Cochrane IBD Group Specialized Register from inception to 21 March 2017. Additionally, we conducted an extensive grey literature search. SELECTION CRITERIA Randomized controlled (placebo, alternative prophylaxis, or no treatment control) trials investigating probiotics (any strain, any dose) for prevention of CDAD, or C. difficile infection were considered for inclusion. DATA COLLECTION AND ANALYSIS Two authors (independently and in duplicate) extracted data and assessed risk of bias. The primary outcome was the incidence of CDAD. Secondary outcomes included detection of C. difficile infection in stool, adverse events, antibiotic-associated diarrhea (AAD) and length of hospital stay. Dichotomous outcomes (e.g. incidence of CDAD) were pooled using a random-effects model to calculate the risk ratio (RR) and corresponding 95% confidence interval (95% CI). We calculated the number needed to treat for an additional beneficial outcome (NNTB) where appropriate. Continuous outcomes (e.g. length of hospital stay) were pooled using a random-effects model to calculate the mean difference and corresponding 95% CI. Sensitivity analyses were conducted to explore the impact of missing data on efficacy and safety outcomes. For the sensitivity analyses, we assumed that the event rate for those participants in the control group who had missing data was the same as the event rate for those participants in the control group who were successfully followed. For the probiotic group, we calculated effects using the following assumed ratios of event rates in those with missing data in comparison to those successfully followed: 1.5:1, 2:1, 3:1, and 5:1. To explore possible explanations for heterogeneity, a priori subgroup analyses were conducted on probiotic species, dose, adult versus pediatric population, and risk of bias as well as a post hoc subgroup analysis on baseline risk of CDAD (low 0% to 2%; moderate 3% to 5%; high > 5%). The overall quality of the evidence supporting each outcome was independently assessed using the GRADE criteria. MAIN RESULTS Thirty-nine studies (9955 participants) met the eligibility requirements for our review. Overall, 27 studies were rated as either high or unclear risk of bias. A complete case analysis (i.e. participants who completed the study) among trials investigating CDAD (31 trials, 8672 participants) suggests that probiotics reduce the risk of CDAD by 60%. The incidence of CDAD was 1.5% (70/4525) in the probiotic group compared to 4.0% (164/4147) in the placebo or no treatment control group (RR 0.40, 95% CI 0.30 to 0.52; GRADE = moderate). Twenty-two of 31 trials had missing CDAD data ranging from 2% to 45%. Our complete case CDAD results proved robust to sensitivity analyses of plausible and worst-plausible assumptions regarding missing outcome data and results were similar whether considering subgroups of trials in adults versus children, inpatients versus outpatients, different probiotic species, lower versus higher doses of probiotics, or studies at high versus low risk of bias. However, in a post hoc analysis, we did observe a subgroup effect with respect to baseline risk of developing CDAD. Trials with a baseline CDAD risk of 0% to 2% and 3% to 5% did not show any difference in risk but trials enrolling participants with a baseline risk of > 5% for developing CDAD demonstrated a large 70% risk reduction (interaction P value = 0.01). Among studies with a baseline risk > 5%, the incidence of CDAD in the probiotic group was 3.1% (43/1370) compared to 11.6% (126/1084) in the control group (13 trials, 2454 participants; RR 0.30, 95% CI 0.21 to 0.42; GRADE = moderate). With respect to detection of C. difficile in the stool pooled complete case results from 15 trials (1214 participants) did not show a reduction in infection rates. C. difficile infection was 15.5% (98/633) in the probiotics group compared to 17.0% (99/581) in the placebo or no treatment control group (RR 0.86, 95% CI 0.67 to 1.10; GRADE = moderate). Adverse events were assessed in 32 studies (8305 participants) and our pooled complete case analysis indicates probiotics reduce the risk of adverse events by 17% (RR 0.83, 95% CI 0.71 to 0.97; GRADE = very low). In both treatment and control groups the most common adverse events included abdominal cramping, nausea, fever, soft stools, flatulence, and taste disturbance. AUTHORS' CONCLUSIONS Based on this systematic review and meta-analysis of 31 randomized controlled trials including 8672 patients, moderate certainty evidence suggests that probiotics are effective for preventing CDAD (NNTB = 42 patients, 95% CI 32 to 58). Our post hoc subgroup analyses to explore heterogeneity indicated that probiotics are effective among trials with a CDAD baseline risk >5% (NNTB = 12; moderate certainty evidence), but not among trials with a baseline risk ≤5% (low to moderate certainty evidence). Although adverse effects were reported among 32 included trials, there were more adverse events among patients in the control groups. The short-term use of probiotics appears to be safe and effective when used along with antibiotics in patients who are not immunocompromised or severely debilitated. Despite the need for further research, hospitalized patients, particularly those at high risk of CDAD, should be informed of the potential benefits and harms of probiotics.
Collapse
Affiliation(s)
| | | | - Lyubov Lytvyn
- McMaster UniversityDepartment of Clinical Epidemiology and BiostatisticsHamiltonCanada
| | - Calvin Ka‐Fung Lo
- The University of British ColumbiaFaculty of MedicineVancouverCanada
| | | | - Dominik Mertz
- McMaster UniversityDivision of Infectious Diseases, Department of MedicineJuravinski Hospital and Cancer Center711 Concession Street, Section G, Level 0, Room 12HamiltonCanadaL8V 1C3
| | - Bradley C Johnston
- Dalhousie UniversityDepartment of Community Health and Epidemiology5790 University AvenueHalifaxCanadaB3H 1V7
| |
Collapse
|
17
|
Zhu XY, Liu F. Probiotics as an adjuvant treatment in Helicobacter pylori eradication therapy. J Dig Dis 2017; 18:195-202. [PMID: 28294543 DOI: 10.1111/1751-2980.12466] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 12/11/2022]
Abstract
Over 80% of individuals infected with Helicobacter pylori (H. pylori) are asymptomatic. Increased resistance to antibiotics and decreased compliance to the therapeutic regimens have led to the failure of eradication therapy. Probiotics, with direct and indirect inhibitory effects on H. pylori in both animal models and clinical trials, have recently been used as a supplementary treatment in H. pylori eradication therapy. Probiotics have been considered useful because of the improvements in H. pylori eradication rates and therapy-related side effects although treatment outcomes using probiotics are controversial due to the heterogeneity of species, strains, doses and therapeutic duration of probiotics. Thus, despite the positive role of probiotics, several factors need to be further considered during their applications. Moreover, adverse events of probiotic use need to be noted. Further investigations into the safety of adjuvant probiotics to H. pylori eradication therapy are required.
Collapse
Affiliation(s)
- Xin Yan Zhu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fei Liu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
An Exploratory Investigation of Endotoxin Levels in Novice Long Distance Triathletes, and the Effects of a Multi-Strain Probiotic/Prebiotic, Antioxidant Intervention. Nutrients 2016; 8:nu8110733. [PMID: 27869661 PMCID: PMC5133117 DOI: 10.3390/nu8110733] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) ischemia during exercise is associated with luminal permeability and increased systemic lipopolysaccharides (LPS). This study aimed to assess the impact of a multistrain pro/prebiotic/antioxidant intervention on endotoxin unit levels and GI permeability in recreational athletes. Thirty healthy participants (25 males, 5 females) were randomly assigned either a multistrain pro/prebiotic/antioxidant (LAB4ANTI; 30 billion CFU·day−1 containing 10 billion CFU·day−1Lactobacillus acidophilus CUL-60 (NCIMB 30157), 10 billion CFU·day−1Lactobacillus acidophillus CUL-21 (NCIMB 30156), 9.5 billion CFU·day−1Bifidobacterium bifidum CUL-20 (NCIMB 30172) and 0.5 billion CFU·day−1Bifidobacterium animalis subspecies lactis CUL-34 (NCIMB 30153)/55.8 mg·day−1 fructooligosaccharides/ 400 mg·day−1 α-lipoic acid, 600 mg·day−1N-acetyl-carnitine); matched pro/prebiotic (LAB4) or placebo (PL) for 12 weeks preceding a long-distance triathlon. Plasma endotoxin units (via Limulus amebocyte lysate chromogenic quantification) and GI permeability (via 5 h urinary lactulose (L): mannitol (M) recovery) were assessed at baseline, pre-race and six days post-race. Endotoxin unit levels were not significantly different between groups at baseline (LAB4ANTI: 8.20 ± 1.60 pg·mL−1; LAB4: 8.92 ± 1.20 pg·mL−1; PL: 9.72 ± 2.42 pg·mL−1). The use of a 12-week LAB4ANTI intervention significantly reduced endotoxin units both pre-race (4.37 ± 0.51 pg·mL−1) and six days post-race (5.18 ± 0.57 pg·mL−1; p = 0.03, ηp2 = 0.35), but only six days post-race with LAB4 (5.01 ± 0.28 pg·mL−1; p = 0.01, ηp2 = 0.43). In contrast, endotoxin units remained unchanged with PL. L:M significantly increased from 0.01 ± 0.01 at baseline to 0.06 ± 0.01 with PL only (p = 0.004, ηp2 = 0.51). Mean race times (h:min:s) were not statistically different between groups despite faster times with both pro/prebiotoic groups (LAB4ANTI: 13:17:07 ± 0:34:48; LAB4: 12:47:13 ± 0:25:06; PL: 14:12:51 ± 0:29:54; p > 0.05). Combined multistrain pro/prebiotic use may reduce endotoxin unit levels, with LAB4ANTI potentially conferring an additive effect via combined GI modulation and antioxidant protection.
Collapse
|
19
|
Inhibitory effects of Lactobacillus fermentum on microbial growth and biofilm formation. Arch Microbiol 2015; 197:1027-32. [PMID: 26267163 DOI: 10.1007/s00203-015-1140-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/17/2015] [Accepted: 08/04/2015] [Indexed: 12/24/2022]
Abstract
Beneficial effects of Lactobacilli have been reported, and lactic bacteria are employed for conservation of foods. Therefore, the effects of a Lactobacillus fermentum strain were analyzed regarding inhibitory effects on staphylococci, Candida albicans and enterotoxigenic enterobacteria by transmission electron microscopy (TEM). TEM of bacterial biofilms was performed using cocultures of bacteriocin-producing L. fermentum 97 with different enterotoxigenic strains: Staphylococcus epidermidis expressing the ica gene responsible for biofilm formation, Staphylococcus aureus producing enterotoxin type A, Citrobacter freundii, Enterobacter cloaceae, Klebsiella oxytoca, Proteus mirabilis producing thermolabile and thermostable enterotoxins determined by elt or est genes, and Candida albicans. L. fermentum 97 changed morphological features and suppressed biofilm formation of staphylococci, enterotoxigenic enterobacteria and Candida albicans; a marked transition to resting states, a degradation of the cell walls and cytoplasm, and a disruption of mature bacterial biofilms were observed, the latter indicating efficiency even in the phase of higher cell density.
Collapse
|
20
|
Kumar PS, Mason MR. Mouthguards: does the indigenous microbiome play a role in maintaining oral health? Front Cell Infect Microbiol 2015; 5:35. [PMID: 26000251 PMCID: PMC4422079 DOI: 10.3389/fcimb.2015.00035] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 03/25/2015] [Indexed: 01/23/2023] Open
Abstract
The existence of symbiotic relationships between bacteria and their hosts in various ecosystems have long been known to science. The human body also hosts vast numbers of bacteria in several habitats. Emerging evidence from the gastro-intestinal tract, genito-urinary tract and respiratory indicates that there are several health benefits to hosting a complex and diverse microbial community. Bacteria colonize the oral cavity within a few minutes after birth and form stable communities. Our knowledge of the oral microbiome has expanded exponentially with development of novel exploratory methods that allow us to examine diversity, structure, function, and topography without the need to cultivate the individual components of the biofilm. The purpose of this perspective, therefore, is to examine the strength of current evidence supporting a role for the oral microbiome in maintaining oral health. While several lines of evidence are emerging to suggest that indigenous oral microbiota may have a role in immune education and preventing pathogen expansion, much more work is needed to definitively establish whether oral bacteria do indeed contribute to sustaining oral health, and if so, the mechanisms underlying this role.
Collapse
Affiliation(s)
- Purnima S. Kumar
- Division of Periodontology, College of Dentistry, The Ohio State UniversityColumbus, OH, USA
| | - Matthew R. Mason
- Division of Biosciences, College of Dentistry, The Ohio State UniversityColumbus, OH, USA
| |
Collapse
|
21
|
Ma Y, Xie TT, Hu Q, Qiu Z, Song F. Sequencing analysis and characterization of the plasmid pBIF10 isolated from Bifidobacterium longum. Can J Microbiol 2015; 61:124-30. [DOI: 10.1139/cjm-2014-0581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A resident plasmid, pBIF10, was isolated from Bifidobacterium longum B200304, and the full-length sequence of pBIF10 was analyzed. In this sequence, we identified at least 17 major open reading frames longer than 200 bp. A tetracycline resistance gene, tetQ, was identified and verified to confer antibiotic resistance to tetracycline. The plasmid replicon with replication protein B gene (repB) and a typical iteron was identified in pBIF10. An artificial clone vector was constructed with the replicon of pBIF10; the results showed that repB controlled plasmid replication in other bifidobacteria host cells at low transformation frequency. Taken together, the analysis and characterization of pBIF10 provided necessary information for the understanding of antibiotic resistance mediated by a plasmid in a Bifidobacterium strain. GC% and repB sequence analyses indicated that pBIF10 was a molecular hybrid of at least 2 other bacterial genera plasmids.
Collapse
Affiliation(s)
- Yongping Ma
- Key Laboratory of Biochemistry and Molecular Biology, Molecular Medical and Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No. 1, Chongqing 400016, People’s Republic of China
| | - Ting-ting Xie
- Key Laboratory of Biochemistry and Molecular Biology, Molecular Medical and Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No. 1, Chongqing 400016, People’s Republic of China
| | - Qiongwen Hu
- Key Laboratory of Biochemistry and Molecular Biology, Molecular Medical and Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No. 1, Chongqing 400016, People’s Republic of China
| | - Zongyin Qiu
- Key Laboratory of Biochemistry and Molecular Biology, Molecular Medical and Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No. 1, Chongqing 400016, People’s Republic of China
| | - Fangzhou Song
- Key Laboratory of Biochemistry and Molecular Biology, Molecular Medical and Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No. 1, Chongqing 400016, People’s Republic of China
| |
Collapse
|
22
|
McFarland LV. Use of probiotics to correct dysbiosis of normal microbiota following disease or disruptive events: a systematic review. BMJ Open 2014; 4:e005047. [PMID: 25157183 PMCID: PMC4156804 DOI: 10.1136/bmjopen-2014-005047] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To assess the evidence for the claim probiotics can correct dysbiosis of the normal microbiota resulting from disease or disruptive events. SETTING Systematic review of published clinical trials of patients receiving a probiotic intervention for the prevention or treatment of various diseases. DATA SOURCES Sources searched (1985-2013): PubMed, EMBASE, Cochrane Database of Systematic Reviews, CINAHL, AMED and ISI Web of Science. Three on-line clinical trial registries were searched: Cochrane Central Register of Controlled trials, MetaRegister of Controlled Trials and National Institutes of Health. REVIEW METHODS Included studies were randomised clinical trials of probiotic interventions having microbiological assays. Studies were evaluated following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines for specific probiotic strains. A standard data extraction form was used to collect the raw data. OUTCOME MEASURES The primary outcome is the degree of microbiota correction by specific probiotic strains. Secondary outcome was the association between the degree of dysbiosis correction and clinical efficacy. RESULTS The review of the literature found three distinct study designs: model A (restoration) assayed patients enrolled with a healthy, undisturbed microbiota and then assayed postdisruptive event and probiotic therapy; model B (alteration) assayed patients with pre-existing disrupted microbiota and then postprobiotic therapy; model C (no dysbiosis) assayed volunteers with no disruptive event prebiotic and postprobiotic. From a total of 63 trials, 83% of the probiotic products using model A restored the microbiota, 56% using model B improved the microbiota and only 21% using model C had any effect on microbiota. Clinical efficacy was more commonly associated with strains capable of restoration of the normal microbiota. CONCLUSIONS The ability to assess the degree of dysbiosis improvement is dependent on the enrolled population and the timing of microbiological assays. The functional claim for correcting dysbiosis is poorly supported for most probiotic strains and requires further research. TRIAL REGISTRATION NUMBER PROSPERO (CRD42014007224).
Collapse
Affiliation(s)
- Lynne V McFarland
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
23
|
Montel MC, Buchin S, Mallet A, Delbes-Paus C, Vuitton DA, Desmasures N, Berthier F. Traditional cheeses: rich and diverse microbiota with associated benefits. Int J Food Microbiol 2014; 177:136-54. [PMID: 24642348 DOI: 10.1016/j.ijfoodmicro.2014.02.019] [Citation(s) in RCA: 356] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 02/17/2014] [Accepted: 02/22/2014] [Indexed: 11/26/2022]
Abstract
The risks and benefits of traditional cheeses, mainly raw milk cheeses, are rarely set out objectively, whence the recurrent confused debate over their pros and cons. This review starts by emphasizing the particularities of the microbiota in traditional cheeses. It then describes the sensory, hygiene, and possible health benefits associated with traditional cheeses. The microbial diversity underlying the benefits of raw milk cheese depends on both the milk microbiota and on traditional practices, including inoculation practices. Traditional know-how from farming to cheese processing helps to maintain both the richness of the microbiota in individual cheeses and the diversity between cheeses throughout processing. All in all more than 400 species of lactic acid bacteria, Gram and catalase-positive bacteria, Gram-negative bacteria, yeasts and moulds have been detected in raw milk. This biodiversity decreases in cheese cores, where a small number of lactic acid bacteria species are numerically dominant, but persists on the cheese surfaces, which harbour numerous species of bacteria, yeasts and moulds. Diversity between cheeses is due particularly to wide variations in the dynamics of the same species in different cheeses. Flavour is more intense and rich in raw milk cheeses than in processed ones. This is mainly because an abundant native microbiota can express in raw milk cheeses, which is not the case in cheeses made from pasteurized or microfiltered milk. Compared to commercial strains, indigenous lactic acid bacteria isolated from milk/cheese, and surface bacteria and yeasts isolated from traditional brines, were associated with more complex volatile profiles and higher scores for some sensorial attributes. The ability of traditional cheeses to combat pathogens is related more to native antipathogenic strains or microbial consortia than to natural non-microbial inhibitor(s) from milk. Quite different native microbiota can protect against Listeria monocytogenes in cheeses (in both core and surface) and on the wooden surfaces of traditional equipment. The inhibition seems to be associated with their qualitative and quantitative composition rather than with their degree of diversity. The inhibitory mechanisms are not well elucidated. Both cross-sectional and cohort studies have evidenced a strong association of raw-milk consumption with protection against allergic/atopic diseases; further studies are needed to determine whether such association extends to traditional raw-milk cheese consumption. In the future, the use of meta-omics methods should help to decipher how traditional cheese ecosystems form and function, opening the way to new methods of risk-benefit management from farm to ripened cheese.
Collapse
Affiliation(s)
| | - Solange Buchin
- INRA, UR342 Technologie et Analyses Laitières, F-39801 Poligny, France
| | - Adrien Mallet
- Normandie Univ, France; UNICAEN, ABTE, F-14032 Caen, France
| | - Céline Delbes-Paus
- INRA, Unité Recherches Fromagères, 20 Côte de Reyne, F-15000 Aurillac, France
| | - Dominique A Vuitton
- UNICAEN, ABTE, F-14032 Caen, France; EA3181/Université de Franche-Comté, 25030, Besançon, France
| | | | | |
Collapse
|
24
|
Govender M, Choonara YE, Kumar P, du Toit LC, van Vuuren S, Pillay V. A review of the advancements in probiotic delivery: Conventional vs. non-conventional formulations for intestinal flora supplementation. AAPS PharmSciTech 2014; 15:29-43. [PMID: 24222267 DOI: 10.1208/s12249-013-0027-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/20/2013] [Indexed: 12/26/2022] Open
Abstract
Probiotic delivery systems are widely used nutraceutical products for the supplementation of natural intestinal flora. These delivery systems vary greatly in effectiveness to exert health benefits for a patient. Probiotic delivery systems can be categorized into conventional, pharmaceutical formulations, and non-conventional, mainly commercial food-based, products. The degree of health benefits provided by these probiotic formulations varies in their ability to deliver viable, functional bacteria in large enough numbers (effectiveness), to provide protection against the harsh effects of the gastric environment and intestinal bile (in vivo protection), and to survive formulation processes (viability). This review discusses the effectiveness of these probiotic delivery systems to deliver viable functional bacteria focusing on the ability to protect the encapsulated probiotics during formulation process as well as against harsh physiological conditions through formulation enhancements using coatings and polymer enhancements. A brief overview on the health benefits of probiotics, current formulation, patient and legal issues facing probiotic delivery, and possible recommendations for the enhanced delivery of probiotic bacteria are also provided. Newer advanced in vitro analyses that can accurately determine the effectiveness of a probiotic formulation are also discussed with an ideal probiotic delivery system hypothesized through a combination of the two probiotic delivery systems described.
Collapse
|
25
|
Cámara M, Gallego-Picó A, Garcinuño R, Fernández-Hernando P, Durand-Alegría J, Sánchez P. An HPLC-DAD method for the simultaneous determination of nine β-lactam antibiotics in ewe milk. Food Chem 2013; 141:829-34. [DOI: 10.1016/j.foodchem.2013.02.131] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 10/24/2012] [Accepted: 02/22/2013] [Indexed: 02/06/2023]
|
26
|
Clinical application of probiotics in the treatment of Helicobacter pylori infection--a brief review. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2013; 47:429-37. [PMID: 23757373 DOI: 10.1016/j.jmii.2013.03.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 03/06/2013] [Accepted: 03/28/2013] [Indexed: 12/13/2022]
Abstract
The role of probiotics in the treatment of gastrointestinal infections is increasingly being documented as an alternative or complement to antibiotics, with the potential to decrease the use of antibiotics or reduce their side effects. Although antibiotics-based Helicobacter pylori eradication treatment is 90% effective, it is expensive and causes antibiotic resistance associated with other adverse effects. Probiotics have an in vitro inhibitory effect on H. pylori. Animal studies demonstrated that probiotic treatment is effective in reducing H. pylori-associated gastric inflammation. About 12 human studies investigated the efficacy of combinations of antibiotics and probiotics, whereas 16 studies used probiotic alone as an alternative to antibiotics for the treatment of H. pylori infection. Most of the studies showed an improvement of H. pylori gastritis and decrease in H. pylori colonization after administration of probiotics. However, no study could demonstrate complete eradication of H. pylori infection by probiotic treatment. Probiotic combinations can reduce adverse effects induced by H. pylori eradication treatment and, thus, have beneficial effects in H. pylori-infected individuals. Long-term intakes of products containing probiotic strains may have a favorable effect on H. pylori infection in humans, particularly by reducing the risk of developing disorders associated with high degrees of gastric inflammation.
Collapse
|
27
|
Goldenberg JZ, Ma SSY, Saxton JD, Martzen MR, Vandvik PO, Thorlund K, Guyatt GH, Johnston BC. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev 2013:CD006095. [PMID: 23728658 DOI: 10.1002/14651858.cd006095.pub3] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Antibiotics are widely prescribed; however they can cause disturbances in gastrointestinal flora which may lead to reduced resistance to pathogens such as Clostridium difficile (C. difficile). Probiotics are live organisms thought to balance the gastrointestinal flora. OBJECTIVES The primary objectives were to assess the efficacy and safety of probiotics for preventing Clostridium difficile-associated diarrhea (CDAD) or C. difficile infection in adults and children. SEARCH METHODS On February 21, 2013 we searched PubMed (1966-2013), EMBASE (1966-2013), Cochrane Central Register of Controlled Trials (The Cochrane Library 2013, Issue 1), CINAHL (1982-2013), AMED (1985-2013), and ISI Web of Science. Additionally, we conducted an extensive grey literature search including contact with industry representatives. SELECTION CRITERIA Randomized controlled (placebo, alternative prophylaxis, or no treatment control) trials investigating probiotics (any strain, any dose) for prevention of CDAD, or C. difficile infection were considered for inclusion. DATA COLLECTION AND ANALYSIS Two authors independently and in duplicate extracted data and assessed risk of bias using pre-constructed, and piloted, data extraction forms. Any disagreements were resolved by a third adjudicator. For articles published in abstract form only, further information was sought by contacting principal authors. The primary outcome was the incidence of CDAD. Secondary outcomes included the incidence of C. difficile infection, adverse events, antibiotic-associated diarrhea (AAD) and length of hospital stay. Dichotomous outcomes (e.g. incidence of CDAD) were pooled using a random-effects model to calculate the relative risk and corresponding 95% confidence interval (95% CI). Continuous outcomes (e.g. length of hospital) were pooled using a random-effects model to calculate the mean difference and corresponding 95% CI. Sensitivity analyses were conducted to explore the impact of missing data on efficacy and safety outcomes. For the sensitivity analyses, we assumed that the event rate for those participants in the control group who had missing data was the same as the event rate for those participants in the control group who were successfully followed. For the probiotic group we calculated effects using the following assumed ratios of event rates in those with missing data in comparison to those successfully followed: 1.5:1, 2:1, 3:1, and 5:1. To explore possible explanations for heterogeneity, a priori subgroup analysis were conducted on probiotic species, dose, adult versus pediatric population, and risk of bias.The overall quality of the evidence supporting each outcome was assessed using the GRADE criteria. MAIN RESULTS A total of 1871 studies were identified with 31 (4492 participants) meeting eligibility requirements for our review. Overall 11 studies were rated as a high risk of bias due mostly to missing outcome data. A complete case analysis (i.e. participants who completed the study) of those trials investigating CDAD (23 trials, 4213 participants) suggests that probiotics significantly reduce this risk by 64%. The incidence of CDAD was 2.0% in the probiotic group compared to 5.5% in the placebo or no treatment control group (RR 0.36; 95% CI 0.26 to 0.51). Sixteen of 23 trials had missing CDAD data ranging from 5% to 45%. These results proved robust to sensitivity analyses of plausible and worst-plausible assumptions regarding missing outcome data and were similar whether considering trials in adults versus children, lower versus higher doses, different probiotic species, or higher versus lower risk of bias. Our judgment is that the overall evidence warrants moderate confidence in this large relative risk reduction. We downgraded the overall quality of evidence for CDAD to 'moderate' due to imprecision. There were few events (154) and the calculated optimal information size (n = 8218) was more than the total sample size. With respect to the incidence of C. difficile infection, a secondary outcome, pooled complete case results from 13 trials (961 participants) did not show a statistically significant reduction. The incidence of C. difficile infection was 12.6% in the probiotics group compared to 12.7% in the placebo or no treatment control group (RR 0.89; 95% CI 0.64 to 1.24). Adverse events were assessed in 26 studies (3964 participants) and our pooled complete case analysis indicates probiotics reduce the risk of adverse events by 20% (RR 0.80; 95% CI 0.68 to 0.95). In both treatment and control groups the most common adverse events included abdominal cramping, nausea, fever, soft stools, flatulence, and taste disturbance. For the short-term use of probiotics in patients that are not immunocompromised or severely debilitated, we consider the strength of this evidence to be moderate. AUTHORS' CONCLUSIONS Based on this systematic review and meta-analysis of 23 randomized controlled trials including 4213 patients, moderate quality evidence suggests that probiotics are both safe and effective for preventing Clostridium difficile-associated diarrhea.
Collapse
|
28
|
|
29
|
Beena Divya J, Kulangara Varsha K, Madhavan Nampoothiri K, Ismail B, Pandey A. Probiotic fermented foods for health benefits. Eng Life Sci 2012. [DOI: 10.1002/elsc.201100179] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
30
|
Ezaki S, Itoh K, Kunikata T, Suzuki K, Sobajima H, Tamura M. Prophylactic probiotics reduce cow's milk protein intolerance in neonates after small intestine surgery and antibiotic treatment presenting symptoms that mimics postoperative infection. Allergol Int 2012; 61:107-13. [PMID: 22377523 DOI: 10.2332/allergolint.11-oa-0305] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 07/13/2011] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND To examine occurrence of cow's milk protein intolerance (CMPI) in newborns that underwent small intestine surgery and the clinical profiles of those newborns with postoperative CMPI, and to evaluate the preventive effects of probiotics on CMPI. METHODS We retrospectively reviewed from 2000 to 2009, a total of 30 newborns required surgery on their small intestines. All of these patients had received antibiotics to prevent postoperative infection. Since 2005 we adopted a protocol of targeted probiotic therapy prophylaxis. RESULTS Eighteen patients received probiotic therapy, while twelve did not. One infant among those eighteen patients and eight patients among those twelve developed CMPI, a significantly lower rate for the group with probiotic therapy than that without it (p < 0.001). Patients with positive cultures for gram positive and gram negative organisms increased in number before and after surgery but then decreased after probiotics treatment. Poor weight gain, gastrointestinal symptoms, and rise in C reactive protein (CRP) levels were observed in all of those nine CMPI patients. Specific IgE antibodies were elevated in four of the nine subjects, and total IgE levels were elevated in seven of them. All CMPI patients had increased level of CRP without proven infections. CONCLUSIONS CMPI was induced in newborns after surgery on their small intestines and antibiotics treatment with presentation of symptoms that mimic postoperative infection. Development of CMPI in this population possibly involves disruption of intestinal flora. Administration of probiotics can reduce the incidence of CMPI after small intestine surgery. The elevated CRP level may be useful in the diagnosis of CMPI.
Collapse
Affiliation(s)
- Shoichi Ezaki
- Division of Neonatal Medicine, Center for Maternal, Fetal and Neonatal Medicine, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan. s_ezaki@saitama−med.ac.jp
| | | | | | | | | | | |
Collapse
|
31
|
Divya JB, Varsha KK, Nampoothiri KM. Newly Isolated Lactic Acid Bacteria with Probiotic Features for Potential Application in Food Industry. Appl Biochem Biotechnol 2012; 167:1314-24. [PMID: 22350936 DOI: 10.1007/s12010-012-9561-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/13/2012] [Indexed: 12/28/2022]
|
32
|
Yin YN, Wang CL, Liu XW, Cui Y, Xie N, Yu QF, Li FJ, Lu FG. Gastric and duodenum microflora analysis after long-term Helicobacter pylori infection in Mongolian Gerbils. Helicobacter 2011; 16:389-97. [PMID: 21923685 DOI: 10.1111/j.1523-5378.2011.00862.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Long-term Helicobacter pylori infection leads to chronic gastritis, peptic ulcer, and gastric malignancies. Indigenous microflora in alimentary tract maintains a colonization barrier against pathogenic microorganisms. This study is aimed to observe the gastric and duodenum microflora alteration after H. pylori infection in Mongolian Gerbils model. MATERIALS AND METHODS A total of 18 Mongolian gerbils were randomly divided into two groups: control group and H. pylori group that were given H. pylori NCTC J99 strain intragastrically. After 12 weeks, H. pylori colonization was identified by rapid urease tests and bacterial culture. Indigenous microorganisms in stomach and duodenum were analyzed by culture method. Histopathologic examination of gastric and duodenum mucosa was also performed. RESULTS Three of eight gerbils had positive H. pylori colonization. After H. pylori infection, Enterococcus spp. and Staphylococcus aureus showed occurrences in stomach and duodenum. Lactobacillus spp. showed a down trend in stomach. The levels and localizations of Bifidobacterium spp., Bacteroides spp., and total aerobes were also modified. Bacteroides spp. significantly increased in H. pylori positive gerbils. No Enterobacteriaceae were detected. Positive colonization gerbils showed a higher histopathologic score of gastritis and a similar score of duodenitis. CONCLUSIONS Long-term H. pylori colonization affected the distribution and numbers of indigenous microflora in stomach and duodenum. Successful colonization caused a more severe gastritis. Gastric microenvironment may be unfit for lactobacilli fertility after long-term H. pylori infection, while enterococci, S. aureus, bifidobacteria, and bacteroides showed their adaptations.
Collapse
Affiliation(s)
- Ya-Ni Yin
- Department of Gastroenterology, Xiangya Second Hospital, Central South University, Changsha City, Hunan Province, China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Shanyinde M, Pickering RM, Weatherall M. Questions asked and answered in pilot and feasibility randomized controlled trials. BMC Med Res Methodol 2011; 11:117. [PMID: 21846349 PMCID: PMC3170294 DOI: 10.1186/1471-2288-11-117] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 08/16/2011] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND In the last decade several authors have reviewed the features of pilot and feasibility studies and advised on the issues that should be addressed within them. We extend this literature by examining published pilot/feasibility trials that incorporate random allocation, examining their stated objectives, results presented and conclusions drawn, and comparing drug and non-drug trials. METHODS A search of EMBASE and MEDLINE databases for 2000 to 2009 revealed 3652 papers that met our search criteria. A random sample of 50 was selected for detailed review. RESULTS Most of the papers focused on efficacy: those reporting drug trials additionally addressed safety/toxicity; while those reporting non-drug trials additionally addressed methodological issues. In only 56% (95% confidence intervals 41% to 70%) were methodological issues discussed in substantial depth, 18% (95% confidence interval 9% to 30%) discussed future trials and only 12% (95% confidence interval 5% to 24%) of authors were actually conducting one. CONCLUSIONS Despite recent advice on topics that can appropriately be described as pilot or feasibility studies the large majority of recently published papers where authors have described their trial as a pilot or addressing feasibility do not primarily address methodological issues preparatory to planning a subsequent study, and this is particularly so for papers reporting drug trials. Many journals remain willing to accept the pilot/feasibility designation for a trial, possibly as an indication of inconclusive results or lack of adequate sample size.
Collapse
Affiliation(s)
- Milensu Shanyinde
- Primary Care and Population Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, UK
| | - Ruth M Pickering
- Primary Care and Population Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, UK
| | - Mark Weatherall
- School of Medicine and Health Sciences, University of Otago Wellington, Wellington, New Zealand
| |
Collapse
|
34
|
Wolvers D, Antoine JM, Myllyluoma E, Schrezenmeir J, Szajewska H, Rijkers GT. Guidance for substantiating the evidence for beneficial effects of probiotics: prevention and management of infections by probiotics. J Nutr 2010; 140:698S-712S. [PMID: 20107143 DOI: 10.3945/jn.109.113753] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The rationale for the use of probiotics in the management of infectious diseases is supported by their potential to influence and stabilize the composition of gut microbiota, enhance colonization resistance, and modulate immune function parameters. A literature review was conducted to determine the efficacy of using probiotics in selected infections: 1) infectious diarrhea in infants and children, 2) traveler's diarrhea, 3) necrotizing enterocolitis in infants, 4) Helicobacter pylori infection, 5) respiratory tract infections in adults and children, 6) ear, nose, and throat infections, and 7) infectious complications in surgical and critically ill patients. The different types of infections that have been subject to clinical studies with different probiotics obviously prevent any generic conclusions. Furthermore, the lack of consistency among studies focusing on 1 specific infection, in study design, applied probiotic strains, outcome parameters, and study population, along with the still limited number of studies, preclude clear and definite conclusions on the efficacy of probiotics and illustrate the need for better-aligned study designs and methodology. Exceptions were the management of infectious diarrhea in infants and traveler's diarrhea, antibiotic-associated diarrhea, and necrotizing enterocolitis. Sufficient consistent data exist for these applications to conclude that certain probiotics, under certain conditions, and in certain target populations, are beneficial in reducing the risk of infection. In addition, some evidence exists, although conclusions are premature, for the management of Helicobacter pylori infection and possible reduction of treatment side effects. Certain probiotics may also reduce the risk of various symptoms of respiratory tract infections in adults and children, including ear, nose, and throat infections, although data are currently far too limited to distill any clinical recommendations in this area. Positive but also negative results have been obtained in prevention of infectious complications in surgical and critically ill patients. For future studies it is recommended that researchers provide adequate power, identify pathogens, and report both clinical outcomes and immune biomarkers relating to putative underlying mechanisms.
Collapse
Affiliation(s)
- Danielle Wolvers
- Unilever Food and Health Research Institute, Vlaardingen, The Netherlands
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Periodontitis is one of the most common chronic inflammatory diseases. A number of putative bacterial pathogens have been associated with the disease and are used as diagnostic markers. In the present study, we compared the prevalence of oral bacterial species in the subgingival biofilm of generalized aggressive periodontitis (GAP) (n = 44) and chronic periodontitis (CP) (n = 46) patients with that of a periodontitis-resistant control group (PR) (n = 21). The control group consisted of subjects at least 65 years of age with only minimal or no periodontitis and no history of periodontal treatment. A total of 555 samples from 111 subjects were included in this study. The samples were analyzed by PCR of 16S rRNA gene fragments and subsequent dot blot hybridization using oligonucleotide probes specific for Aggregatibacter (Actinobacillus) actinomycetemcomitans, Porphyromonas gingivalis, Prevotella intermedia, Tannerella forsythia, a Treponema denticola-like phylogroup (Treponema phylogroup II), Treponema lecithinolyticum, Campylobacter rectus, Fusobacterium spp., and Fusobacterium nucleatum, as well as Capnocytophaga ochracea. Our data confirm a high prevalence of the putative periodontal pathogens P. gingivalis, P. intermedia, and T. forsythia in the periodontitis groups. However, these species were also frequently detected in the PR group. For most of the species tested, the prevalence was more associated with increased probing depth than with the subject group. T. lecithinolyticum was the only periodontopathogenic species showing significant differences both between GAP and CP patients and between GAP patients and PR subjects. C. ochracea was associated with the PR subjects, regardless of the probing depth. These results indicate that T. lecithinolyticum may be a diagnostic marker for GAP and C. ochracea for periodontal health. They also suggest that current presumptions of the association of specific bacteria with periodontal health and disease require further evaluation.
Collapse
|
36
|
Kaur IP, Kuhad A, Garg A, Chopra K. Probiotics: Delineation of Prophylactic and Therapeutic Benefits. J Med Food 2009; 12:219-35. [DOI: 10.1089/jmf.2007.0544] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Indu Pal Kaur
- Division of Pharmaceutics, University Institute of Pharmaceutical Sciences, UGC Center for Advanced Studies, Panjab University, Chandigarh, India
| | - Anurag Kuhad
- Division of Pharmacology, University Institute of Pharmaceutical Sciences, UGC Center for Advanced Studies, Panjab University, Chandigarh, India
| | - Amita Garg
- Division of Pharmaceutics, University Institute of Pharmaceutical Sciences, UGC Center for Advanced Studies, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- Division of Pharmacology, University Institute of Pharmaceutical Sciences, UGC Center for Advanced Studies, Panjab University, Chandigarh, India
| |
Collapse
|
37
|
Williams EA, Stimpson J, Wang D, Plummer S, Garaiova I, Barker ME, Corfe BM. Clinical trial: a multistrain probiotic preparation significantly reduces symptoms of irritable bowel syndrome in a double-blind placebo-controlled study. Aliment Pharmacol Ther 2009; 29:97-103. [PMID: 18785988 DOI: 10.1111/j.1365-2036.2008.03848.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The efficacy of probiotics in alleviating the symptoms of irritable bowel syndrome (IBS) appears to be both strain- and dose-related. AIM To investigate the effect of LAB4, a multistrain probiotic preparation on symptoms of IBS. This probiotic preparation has not previously been assessed in IBS. METHODS Fifty-two participants with IBS, as defined by the Rome II criteria, participated in this double blind, randomized, placebo-controlled study. Participants were randomized to receive either a probiotic preparation comprising two strains of Lactobacillus acidophilus CUL60 (NCIMB 30157) and CUL21 (NCIMB 30156), Bifidobacterium lactis CUL34 (NCIMB 30172) and Bifidobacterium bifidum CUL20 (NCIMB 30153) at a total of 2.5 × 10(10) cfu/capsule or a placebo for 8 weeks. Participants reported their IBS symptoms using a questionnaire fortnightly during the intervention and at 2 weeks post-intervention. RESULTS A significantly greater improvement in the Symptom Severity Score of IBS and in scores for quality of life, days with pain and satisfaction with bowel habit was observed over the 8-week intervention period in the volunteers receiving the probiotic preparation than in the placebo group. CONCLUSION LAB4 multistrain probiotic supplement may benefit subjects with IBS.
Collapse
Affiliation(s)
- E A Williams
- Human Nutrition Unit, School of Medicine, The University of Sheffield, Sheffield, UK.
| | | | | | | | | | | | | |
Collapse
|
38
|
Role of probiotics in antibiotic-associated diarrhea, Clostridium difficile-associated diarrhea, and recurrent Clostridium difficile-associated diarrhea. J Clin Gastroenterol 2008; 42 Suppl 2:S64-70. [PMID: 18545161 DOI: 10.1097/mcg.0b013e3181646d09] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The role of probiotics in the prevention and treatment of antibiotic-associated diarrhea, Clostridium difficile diarrhea, and recurrent C. difficile diarrhea is reviewed. Various probiotics have variable efficacy. More studies are needed to define further their efficacies, roles, and indications.
Collapse
|
39
|
Zuccotti GV, Meneghin F, Raimondi C, Dilillo D, Agostoni C, Riva E, Giovannini M. Probiotics in clinical practice: an overview. J Int Med Res 2008; 36 Suppl 1:1A-53A. [PMID: 18230282 DOI: 10.1177/14732300080360s101] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The observation that intestinal bacterial microflora might be able to influence immune system surveillance through changed nutritional habits has raised awareness of the role of probiotics. These are live microorganisms that are able to reach the gastrointestinal tract and alter its microfloral composition, producing beneficial health effects when consumed in adequate amounts. Recent clinical trials have evaluated the clinical effectiveness of probiotics in the treatment and prevention of a wide range of acute and chronic gastrointestinal diseases, and also non-gastrointestinal diseases, such as atopy, respiratory infections, vaginitis and hypercholesterolaemia. Probiotic supplements are generally regarded as safe because the microorganisms they contain are identical to those found in human gastrointestinal and vaginal microflora. Guidelines on the use of probiotics in the clinical setting require periodical updates for the latest data to be included in clinical applications. The purpose of this clinical report is to review current evidence on the use of probiotics in a variety of gastrointestinal and non-gastrointestinal conditions.
Collapse
Affiliation(s)
- G V Zuccotti
- Department of Paediatrics, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel disease largely affecting low birth weight, premature infants. Once acquired, NEC is accompanied by significant mortality and morbid sequelae. Our understanding of the pathophysiology of NEC continues to evolve, and the development of NEC is likely multifactorial with resultant bowel injury mediated through a final, common inflammatory pathway. The predisposition for NEC appears to involve the interplay between intestinal integrity and function, enteral feeding and bacterial colonization, and regulation of the gastrointestinal and systemic inflammatory response. Commensal organisms or probiotics have been shown to be crucial in the development and modulation of each of these factors within the intestinal epithelium. As a result, probiotic supplementation has been proposed as a promising new intervention for the prevention of NEC. To understand the potential utility of probiotics in NEC, we will discuss: the components of gut defense; the role of the intestinal ecosystem in modulating immunity and inflammation; bacterial colonization patterns in the preterm infant compared with patterns seen in the healthy, full-term infant; the evidence for probiotic use in other populations and diseases; and finally, the evidence of probiotic use specific to the preterm infant and NEC.
Collapse
Affiliation(s)
- Camilia R Martin
- Department of Pediatrics, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | |
Collapse
|
41
|
Koning CJM, Jonkers DMAE, Stobberingh EE, Mulder L, Rombouts FM, Stockbrügger RW. The effect of a multispecies probiotic on the intestinal microbiota and bowel movements in healthy volunteers taking the antibiotic amoxycillin. Am J Gastroenterol 2008; 103:178-89. [PMID: 17900321 DOI: 10.1111/j.1572-0241.2007.01547.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND One of the side effects of antimicrobial therapy is a disturbance of the intestinal microbiota potentially resulting in antibiotic-associated diarrhea (AAD). In this placebo-controlled double-blind study, the effect of a multispecies probiotic on the composition and metabolic activity of the intestinal microbiota and bowel habits was studied in healthy volunteers taking amoxycillin. METHODS Forty-one healthy volunteers were given 500 mg amoxycillin twice daily for 7 days and were randomized to either 5 g of a multispecies probiotic, Ecologic AAD (10(9) cfu/g), or placebo, twice daily for 14 days. Feces and questionnaires were collected on day 0, 7, 14, and 63. Feces was analyzed as to the composition of the intestinal microbiota, and beta-glucosidase activity, endotoxin concentration, Clostridium difficile toxin A, short chain fatty acids (SCFAs), and pH were determined. Bowel movements were scored according to the Bristol stool form scale. RESULTS Mean number of enterococci increased significantly from log 4.1 at day 0 to log 5.8 (day 7) and log 6.9 (day 14) cfu/g feces (P < 0.05) during probiotic intake. Although no other significant differences were observed between both intervention groups, within each group significant changes were found over time in both microbial composition and metabolic activity. Moreover, bowel movements with a frequency >or=3 per day for at least 2 days and/or a consistency >or=5 for at least 2 days were reported less frequently in the probiotic compared to the placebo group (48%vs 79%, P < 0.05). CONCLUSIONS Apart from an increase in enterococci no significant differences in microbial composition and metabolic activity were observed in the probiotic compared with the placebo group. However, changes over time were present in both groups, which differed significantly between the probiotic and the placebo arm, suggesting that the amoxycillin effect was modulated by probiotic intake. Moreover, the intake of a multispecies probiotic significantly reduced diarrhea-like bowel movements in healthy volunteers receiving amoxycillin.
Collapse
Affiliation(s)
- Catherina J M Koning
- Division of Gastroenterology-Hepatology, University Hospital Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
42
|
Bertrand X, Dufour V, Millon L, Beuvier E, Gbaguidi-Haore H, Piarroux R, Vuitton DA, Talon D. Effect of cheese consumption on emergence of antimicrobial resistance in the intestinal microflora induced by a short course of amoxicillin-clavulanic acid. J Appl Microbiol 2007; 102:1052-9. [PMID: 17381749 DOI: 10.1111/j.1365-2672.2006.03148.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AIM To study in a sequential prospective trial, the effect of cheese consumption on the emergence of Escherichia coli and enterococci resistance to amoxicillin after amoxicillin-clavulanic acid (amoxiclav) treatment. METHODS AND RESULTS The study comprised two phases separated by 1 year. Each phase lasted 75 days for each volunteer (from day -13 to day 61). During the first phase, 18 healthy volunteers were given a 1-g dose of amoxiclav orally twice a day for 5 days (from day 0 to day 4). The design of phase 2 was identical to that of phase 1, except that the volunteers consumed 100 g of hard-cooked cheese from day -6 to day 19. Faecal samples were collected 20 times throughout the trial and were quantitatively assayed for total and amoxicillin-resistant (Amox(R)) E. coli and enterococci. The consumption of experimental cheese was associated with a decrease of Amox(R) enterococci during the post-antibiotic period, with the maximum level of Amox(R) enterococci falling from 6.2% to 0.03%. This effect was not observed for E. coli, and the type of cheese (raw milk vs pasteurized milk) did not influence the results. CONCLUSIONS Consumption of cheese during amoxiclav treatment reduces the emergence of Amox(R) enterococci in faeces. SIGNIFICANCE AND CLINICAL IMPACT OF THE STUDY: Our clinical pilot trial suggests that there are likely to be benefits from consuming probiotic-containing cheese during antibiotic treatment.
Collapse
Affiliation(s)
- X Bertrand
- Service d'Hygiène Hospitalière, Centre Hospitalier Universitaire de Besançon, Besançon, France.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Tong JL, Ran ZH, Shen J, Zhang CX, Xiao SD. Meta-analysis: the effect of supplementation with probiotics on eradication rates and adverse events during Helicobacter pylori eradication therapy. Aliment Pharmacol Ther 2007; 25:155-68. [PMID: 17229240 DOI: 10.1111/j.1365-2036.2006.03179.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Recent evidence found probiotics could inhibit Helicobacter pylori colonization from both in vitro and in vivo studies. AIM To systematically evaluate whether adding probiotics to anti-H. pylori regimens could improve eradication rates and reduce side effects during anti-H. pylori treatment. METHODS Eligible articles were identified by searches of electronic databases. We included all randomized trials comparing probiotics supplementation to placebo or no treatment during anti-H. pylori regimens. Statistical analysis was performed with Review Manager 4.2.8. Subanalysis/Sensitivity analysis was also performed. RESULTS We identified 14 randomized trials (n = 1671). Pooled H. pylori eradication rates were 83.6% (95% CI = 80.5-86.7%) and 74.8% (95% CI = 71.1-78.5%) for patients with or without probiotics by intention-to-treat analysis, respectively, the odds ratio (OR) was 1.84 (95% CI = 1.34-2.54); the occurrence of total side effects were 24.7% (95% CI = 20.0-29.4%) and 38.5% (95% CI = 33.0-44.1%) for groups with or without probiotics, especially for diarrhoea, the summary OR was 0.44 (95% CI = 0.30-0.66). CONCLUSIONS Our review suggests that supplementation with probiotics could be effective in increasing eradication rates of anti-H. pylori therapy, and could be considered helpful for patients with eradication failure. Furthermore, probiotics show a positive impact on H. pylori therapy-related side effects.
Collapse
Affiliation(s)
- J L Tong
- Department of Gastroenterology, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
44
|
Saarela M, Maukonen J, von Wright A, Vilpponen-Salmela T, Patterson AJ, Scott KP, Hämynen H, Mättö J. Tetracycline susceptibility of the ingested Lactobacillus acidophilus LaCH-5 and Bifidobacterium animalis subsp. lactis Bb-12 strains during antibiotic/probiotic intervention. Int J Antimicrob Agents 2007; 29:271-80. [PMID: 17207972 DOI: 10.1016/j.ijantimicag.2006.09.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 09/26/2006] [Accepted: 09/26/2006] [Indexed: 01/07/2023]
Abstract
We investigated the effects of oral therapy with doxycycline, a tetracycline group antibiotic, on the gastrointestinal (GI) survival and tetracycline susceptibility of probiotic strains Lactobacillus acidophilus LaCH-5 and Bifidobacterium animalis subsp. lactis Bb-12. In addition, the influence of doxycycline therapy on the diversity of the predominant faecal microbiota was evaluated by polymerase chain reaction-denaturing gradient gel electrophoresis (PCR-DGGE). Faecal samples from the antibiotic group (receiving antibiotics and probiotics) and the control group (receiving probiotics only) were analysed for anaerobically and aerobically growing bacteria, bifidobacteria and lactic acid bacteria as well as for the dominant microbiota. Although doxycycline consumption did not have a large impact on GI survival of the probiotics, it had a detrimental effect on the bifidobacteria and on the diversity of the dominant faecal microbiota. A higher proportion of tetracycline-resistant anaerobically growing bacteria and bifidobacteria was detected in the antibiotic group than in the control group. Several antibiotic group subjects had faecal B. animalis subsp. lactis Bb-12-like isolates with reduced tetracycline susceptibility. This was unlikely to be due to the acquisition of novel tetracycline resistance determinants, since only tet(W), which is also present in the ingested B. animalis subsp. lactis Bb-12, was found in the resistant isolates. Thus, concomitant ingestion of probiotic L. acidophilus LaCH-5 and B. animalis subsp. lactis Bb-12 with the antibiotic did not generate a safety risk regarding the possible GI transfer of tetracycline resistance genes to the ingested strains.
Collapse
Affiliation(s)
- Maria Saarela
- VTT, Biotechnology, P.O. Box 1000, FI-02044 VTT, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Myllyluoma E, Ahlroos T, Veijola L, Rautelin H, Tynkkynen S, Korpela R. Effects of anti-Helicobacter pylori treatment and probiotic supplementation on intestinal microbiota. Int J Antimicrob Agents 2006; 29:66-72. [PMID: 17141481 DOI: 10.1016/j.ijantimicag.2006.08.034] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Revised: 08/21/2006] [Accepted: 08/21/2006] [Indexed: 01/15/2023]
Abstract
The aims of this study were (i) to evaluate the effect of recommended antimicrobial treatment of Helicobacter pylori infection, consisting of clarithromycin, amoxicillin and lansoprazole, on intestinal microbiota and (ii) to determine the ability of a probiotic combination containing Lactobacillus rhamnosus GG, L. rhamnosus LC705, Propionibacterium freudenreichii ssp. shermanii JS and Bifidobacterium breve Bb99 to prevent treatment-induced alterations in the intestinal microbiota. Faecal samples were obtained from 39 H. pylori-infected patients randomised into two treatment groups. In addition, 19 H. pylori-negative volunteers were included in the study as a control group. Samples were collected before, during and after treatment and microbiota were analysed by fluorescence in situ hybridisation and culture. The quantities of the predominant bacterial groups were altered significantly in both groups and disturbances were seen even 9 weeks after treatment was complete. Probiotics slightly counteracted the effects of anti-H. pylori treatment, seen as significantly less alterations in the total numbers of aerobes and lactobacilli/enterococci. At baseline, the composition of the microbiota between H. pylori-positive versus H. pylori-negative control individuals differed with regard to clostridia and the total number of anaerobes. The recommended treatment for H. pylori infection induces long-term disturbances in the intestinal microbiota. The probiotic combination appeared to result in only minor changes in the microbiota.
Collapse
Affiliation(s)
- E Myllyluoma
- Institute of Biomedicine, Pharmacology, University of Helsinki, Helsinki, Finland; Foundation for Nutrition Research, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
46
|
Kumar PS, Leys EJ, Bryk JM, Martinez FJ, Moeschberger ML, Griffen AL. Changes in periodontal health status are associated with bacterial community shifts as assessed by quantitative 16S cloning and sequencing. J Clin Microbiol 2006; 44:3665-73. [PMID: 17021095 PMCID: PMC1594761 DOI: 10.1128/jcm.00317-06] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The gingival sulcus contains a complex ecosystem that includes many uncultivated bacteria. Understanding the dynamics of this ecosystem in transitions between health and disease is important in advancing our understanding of the bacterial etiology of periodontitis. The objective of this longitudinal study was to examine the stability of bacterial colonization in the gingival crevice and to explore the relationship between shifts in microbial composition and changes in periodontal health status using a comprehensive, quantitative, culture-independent approach. Subgingival plaque samples and periodontal data were collected from 24 subjects over 2 years. Baseline and 2-year plaque samples were analyzed using quantitative ribosomal 16S cloning and sequencing. Ten subjects remained periodontally healthy over 2 years, the periodontal health of seven subjects worsened, and seven subjects showed clinical improvement. Bacterial stability was greatest among healthy, clinically stable subjects and lowest for subjects whose periodontal status worsened (P = 0.01). Higher numbers of species lost or gained were also observed for subjects whose clinical status changed (P = 0.009). This provides evidence that a change in periodontal status is accompanied by shifts within the bacterial community. Based on these data, measures of microbial stability may be useful in clinical diagnosis and prognosis. Regarding individual species, increases in levels of the uncultivated phylotype Veillonella sp. oral clone X042, a gram-negative bacterium and the most common member of the subgingival bacterial community, were associated with periodontal health (P = 0.04), suggesting that this is an important beneficial species. Filifactor alocis, a gram-positive anaerobe, was found at higher levels in subjects with disease (P = 0.01).
Collapse
Affiliation(s)
- Purnima S Kumar
- The Ohio State University, Section of Pediatric Dentistry, 305 W. 12th Ave., P.O. Box 182357, Columbus, OH 43218, USA
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW A disturbance in microbial balance of the gastrointestinal tract is often associated with diarrhea. Therefore, probiotics, as beneficial microorganisms for host health, have attracted clinical attention for their potential therapeutic application in the treatment of diarrhea. This review focuses on new research findings relevant to the effects of probiotics on diarrhea prevention and treatment and potential mechanisms of action for this alternative therapy for diarrhea. RECENT FINDINGS Clinical trials suggest potential beneficial effects of probiotic therapy for preventing and treating antibiotic-associated diarrhea, acute diarrhea including rotavirus-induced diarrhea, traveler's diarrhea, and diarrhea-predominant irritable bowel syndrome. The most extensively studied probiotics for diarrhea are Lactobacillus, Bifidobacterium and Saccharomyces, with potential mechanisms of therapeutic action based on the protection of intestinal epithelial cell and barrier function, prevention of enterotoxin binding to intestinal epithelial cells, and regulation of intestinal microbial environment. SUMMARY Growing evidence suggests that probiotics may serve as a functional food in the treatment of diarrhea. Remaining challenges include identifying mechanisms of action to provide the basis of more refined hypothesis-driven clinical trials. The correct combination and concentration of probiotics applied to the appropriate gastrointestinal disorders may improve the efficacy of this approach for diarrhea and other diseases.
Collapse
Affiliation(s)
- Fang Yan
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | |
Collapse
|
48
|
Brunser O, Gotteland M, Cruchet S, Figueroa G, Garrido D, Steenhout P. Effect of a milk formula with prebiotics on the intestinal microbiota of infants after an antibiotic treatment. Pediatr Res 2006; 59:451-6. [PMID: 16492988 DOI: 10.1203/01.pdr.0000198773.40937.61] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibiotics exert deleterious effects on the intestinal microbiota, favoring the emergence of opportunistic bacteria and diarrhea. Prebiotics are nondigestible food components that stimulate the growth of bifidobacteria. Our aim was to evaluate the effects on the intestinal microbiota of a prebiotic-supplemented milk formula after an antibiotic treatment. A randomized, double-blind, controlled clinical trial was carried out in 140 infants 1-2 y of age distributed into two groups after a 1-wk amoxicillin treatment (50 mg/kg/d) for acute bronchitis. The children received for 3 wk >500 mL/d of a formula with prebiotics (4.5 g/L) or a control without prebiotics. Fecal samples were obtained on d -7 (at the beginning of the antibiotic treatment), on d 0 (end of the treatment and before formula administration), and on d 7 and 21 (during formula administration). Counts of Bifidobacterium, Lactobacillus-Enterococcus, Clostridium lituseburiense cluster, Clostridium histolyticum cluster, Escherichia coli, and Bacteroides-Prevotella were evaluated by fluorescent in situ hybridization (FISH) and flow cytometry. Tolerance and gastrointestinal symptoms were recorded daily. Amoxicillin decreased total fecal bacteria and increased E. coli. The prebiotic significantly increased bifidobacteria from 8.17 +/- 1.46 on d 0 to 8.54 +/- 1.20 on d 7 compared with the control 8.22 +/- 1.24 on d 0 versus 7.95 +/- 1.54 on d 7. The Lactobacillus population showed a similar tendency while the other bacteria were unaffected. No gastrointestinal symptoms were detected during the prebiotic administration. Prebiotics in a milk formula increase fecal bifidobacteria early after amoxicillin treatment without inducing gastrointestinal symptoms.
Collapse
Affiliation(s)
- Oscar Brunser
- Laboratory of Ultrastructure, Institute of Nutrition and Food Technology (INTA), University of Chile, Chile.
| | | | | | | | | | | |
Collapse
|
49
|
|