1
|
Yuzefpolskaya M, Bohn B, Ladanyi A, Khoruts A, Colombo PC, Demmer RT. Oral and gut microbiome alterations in heart failure: Epidemiology, pathogenesis and response to advanced heart failure therapies. J Heart Lung Transplant 2023; 42:291-300. [PMID: 36586790 DOI: 10.1016/j.healun.2022.12.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/18/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Despite significant advances in therapies, heart failure (HF) remains a progressive disease that, once advanced, is associated with significant death and disability. Cardiac replacement therapies with left ventricular assist device (LVAD) and heart transplantation (HT) are the only treatment options for advanced HF, while lifesaving they can also be lifespan limiting due to the associated complications. Systemic inflammation is mechanistically important in HF pathophysiology and progression. However, directly targeting inflammation in HF has not been beneficial thus far. These failed attempts at therapeutics might be related to our limited understanding of the factors that cause inflammation in HF, and, therefore, to our inability to investigate these triggers in interventional studies. Observational studies have consistently demonstrated associations between alterations in the digestive (gut and oral) microbiome, inflammation and HF risk and progression. Additionally, recent data indicate that these microbial perturbations persist following LVAD and HT, along with residual inflammation and oxidative stress. Furthermore, there is rising recognition of the critical contribution of the microbiome to the metabolism of immunosuppressive drugs after HT. Cumulatively, these findings might posit a mechanistic link between microbiome alterations, systemic inflammation, and adverse outcomes in HF patients before and after cardiac replacement therapies. This review (1) provides an update on available data linking changes in digestive tract microbiota, inflammation, and oxidative stress, to HF pathogenesis and progression; (2) describes evolution of these relationships following LVAD and HT; and (3) outlines present and future intervention strategies that can manipulate the microbiome and possibly modify HF disease trajectory.
Collapse
Affiliation(s)
- Melana Yuzefpolskaya
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York.
| | - Bruno Bohn
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Annamaria Ladanyi
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine University of Minnesota, Minneapolis, Minnesota
| | - Paolo C Colombo
- Division of Cardiovascular Medicine, Columbia University Irving Medical Center, New York City, New York
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota; Division of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
2
|
Li Y, Chen M, Ma Y, Yang Y, Cheng Y, Ma H, Ren D, Chen P. Regulation of viable/inactivated/lysed probiotic Lactobacillus plantarum H6 on intestinal microbiota and metabolites in hypercholesterolemic mice. NPJ Sci Food 2022; 6:50. [PMID: 36316361 PMCID: PMC9622822 DOI: 10.1038/s41538-022-00167-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/11/2022] [Indexed: 01/24/2023] Open
Abstract
Evidence suggests that probiotic interventions reduce non-communicable diseases (NCDs) risk. However, its therapeutic effect and mechanism are still unclear. To evaluate the hypocholesterolemic effect of Lactobacillus plantarum H6 (L.p H6), a new commercial patent strain capable of preventing hypercholesterolemia, and its mechanism in depth, three states of the strain were prepared, namely, viable (vH6), heat-inactivated (iH6), and ultrasonically-lysed (uH6) bacteria cells. The results showed that v/i/uH6 cells could lower serum and liver blood lipid levels, alleviate liver damage and improve glucose tolerance test (GTT) and insulin tolerance test (ITT) indexes. v/i/uH6 cells improved the gut microbial composition and significantly reduced the Firmicutes to Bacteroidetes ratio (F/B ratio) in feces. In particular, Muribaculaceae may be a potential biomarker for effective cholesterol reduction. Also, the recovery of these biochemical indices and gut microbiome was found following fecal microbiota transplantation (FMT) using stool from vH6 treated mice. The v/i/uH6 cells increased the intestinal flora metabolism of vitamins-cofactors, as well as amino acids, while decreasing the relative content of primary bile acids. The Pearson correlation analysis showed that norank_f__Muribaculaceae and Lactobacillus had a negative correlation with blood lipid levels. Overall, v/i/uH6 cells were effective in improving hypercholesterolemia in mice, and this effect was attributed partly to the regulation of intestinal microbiota and metabolites related to lipid metabolism. Our findings provided a theoretical basis for the industrial development of probiotics and postbiotics and the treatment of cholesterol diseases.
Collapse
Affiliation(s)
- Yue Li
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| | - Mengling Chen
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| | - Yuxuan Ma
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| | - Yue Yang
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| | - Ying Cheng
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| | - Huijing Ma
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| | - Dayong Ren
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| | - Ping Chen
- grid.464353.30000 0000 9888 756XCollege of Food Science and Engineering, Jilin Agricultural University, 130118 Changchun, China
| |
Collapse
|
3
|
Panigrahi MK, Kaliaperumal V, Akella A, Venugopal G, Ramadass B. Mapping microbiome-redox spectrum and evaluating Microbial-Redox Index in chronic gastritis. Sci Rep 2022; 12:8450. [PMID: 35589904 PMCID: PMC9120160 DOI: 10.1038/s41598-022-12431-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/11/2022] [Indexed: 12/27/2022] Open
Abstract
Peptic ulcer disease (PUD) and chronic gastritis are prevalent in developing countries. The role of oxidative stress in the pathogenesis of gastrointestinal mucosal disorders is well recognized. In PUD, the gastric mucosa and its associated microbiome are subject to diet and stress-induced oxidative perturbations. Tissue redox potential (ORP) measurement can quantify oxidative stress, reflecting the balance between prooxidants and antioxidants. This study hypothesizes that the oxidative stress quantified by tissue ORP will be associated with characteristic changes in the mucosa-associated microbiome in PUD and gastritis. In addition, we propose using relative microbial abundance as a quantitative marker of mucosal health. Endoscopy was performed to obtain gastric mucosal biopsies from ten PUD and ten non-ulcer dyspepsia (NUD) patients. The tissue ORP was measured directly with a microelectrode using a biopsy specimen. A second specimen from an adjacent site was subjected to 16s rRNA gene sequencing. From the OTUs, the relative abundance of the microbial taxon in each of the samples was derived. We analyzed the genome of the predominant species for genes encoding the utilization of oxygen as an electron acceptor in respiration and for the presence of antioxidant defense mechanisms. The organisms were then grouped based on their established and inferred redox traits. Shannon diversity index and Species richness were calculated on rarefied data. The relative abundance of organisms that prefer high ORP over those that favor low ORP is conceived as the “Microbial Redox Index (MRI),” an indicator of mucosal health. In the gastric mucosa, aerobic species predominate and are more diverse than the anaerobes. The predominant aerobes are Helicobacter pylori and Sphingobacterium mizutaii. The abundance of these two species had an inverse correlation with the abundance of low ORP preferring anaerobes. Their relative abundance ratio (Microbial Redox Index) correlated with the tissue oxidation–reduction potential (ORP), a direct measure of oxidative stress. Correlation analysis also revealed that the abundance of all anaerobes inversely correlated with the dominant aerobic taxa. In addition, Shannon and Species richness diversity indices, the probable indicators of mucosal health, were negatively correlated with Microbial Redox Index. Using PUD as a prototype mucosal disease, this article describes a generalized approach to infer and quantify mucosal oxidative stress by analyzing the relative abundance of microorganisms that preferentially grow at the extremes of the tissue redox potential. This ratiometric Microbial Redox Index can also be assessed using simple qPCR without the need for sequencing. The approach described herein may be helpful as a widely applicable quantitative measure of mucosal health with prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Manas Kumar Panigrahi
- Department of Gastroenterology, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Venkatesh Kaliaperumal
- MYAS-NIN Department of Sports Science, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Abhishek Akella
- Center of Excellence for Clinical Microbiome Research, All India Institute of Medical Sciences, Bhubaneswar, 751019, India
| | - Giriprasad Venugopal
- Center of Excellence for Clinical Microbiome Research, All India Institute of Medical Sciences, Bhubaneswar, 751019, India
| | - Balamurugan Ramadass
- Center of Excellence for Clinical Microbiome Research, All India Institute of Medical Sciences, Bhubaneswar, 751019, India. .,Department of Biochemistry, All India Institute of Medical Sciences, Bhubaneswar, India.
| |
Collapse
|
4
|
Kushak RI, Sengupta A, Winter HS. Interactions between the intestinal microbiota and epigenome in individuals with autism spectrum disorder. Dev Med Child Neurol 2022; 64:296-304. [PMID: 34523735 DOI: 10.1111/dmcn.15052] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/27/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by variable impairment of cognitive function and interpersonal relationships. Furthermore, some individuals with ASD have gastrointestinal disorders that have been correlated with impairments in intestinal microbiota. Gut microbiota are important not only for intestinal health, but also for many other functions including food digestion, energy production, immune system regulation, and, according to current data, behavior. Disruption of the indigenous microbiota, microbial dysbiosis (imbalance between microorganisms present in the gut), overgrowth of potentially pathogenic microorganisms, a less diverse microbiome, or lower levels of beneficial bacteria in children with ASD can affect behavior. Metabolome analysis in children with ASD has identified perturbations in multiple metabolic pathways that might be associated with cognitive functions. Recent studies have shown that the intestinal microbiome provides environmental signals that can modify host response to stimuli by modifying the host epigenome, which affects DNA methylation, histone modification, and non-coding RNAs. The most studied microbiota-produced epigenetic modifiers are short-chain fatty acids, although other products of intestinal microbiota might also cause epigenetic modifications in the host's DNA. Here we review evidence suggesting that epigenetic alterations caused by modification of gene expression play an important role in understanding ASD.
Collapse
Affiliation(s)
- Rafail I Kushak
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ashok Sengupta
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Harland S Winter
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Beeck R, Glöckl G, Krause J, Schick P, Weitschies W. Mimicking the dynamic Colonic microbiota in vitro to gain a better understanding on the in vivo metabolism of xenobiotics: Degradation of sulfasalazine. Int J Pharm 2021; 603:120704. [PMID: 33991596 DOI: 10.1016/j.ijpharm.2021.120704] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 12/28/2022]
Abstract
Due to the potential effects of colonic metabolism, the interest in the composition and action of intestinal microbiota has increased significantly throughout the last 10 years. Recently focus is turning to the development and implementation of in vitro tools closely simulating in vivo colonic metabolic processes suitable for routine use. The aim of the present study is to compare the metabolization of the model drug sulfasalazine utilizing the novel dynamic bioreactor MimiCol and a standard static batch fermenter inoculated with cryopreserved faecal microbiota. Major advantages of the novel bioreactor MimiCol are the smaller media volume which is closer to in vivo conditions, the possibility to perform media changes and the closer simulation of in vivo mixing patterns. The study proved that the MimiCol is able to simulate the dynamic conditions found within the ascending colon. The dynamic conditions within the MimiCol led to an almost 2-fold increase of the metabolization rate constant in comparison to the static batch fermenter. Our study was able to prove that the novel dynamic bioreactor MimiCol is able to closely simulate physiologically relevant conditions.
Collapse
Affiliation(s)
- Regine Beeck
- University of Greifswald, Institute of Pharmacy, Center of Drug Absorption and Transport (C_DAT), Felix-Hausdorff-Str. 3, D-17489 Greifswald, Germany.
| | - Gunnar Glöckl
- University of Greifswald, Institute of Pharmacy, Center of Drug Absorption and Transport (C_DAT), Felix-Hausdorff-Str. 3, D-17489 Greifswald, Germany.
| | - Julius Krause
- University of Greifswald, Institute of Pharmacy, Center of Drug Absorption and Transport (C_DAT), Felix-Hausdorff-Str. 3, D-17489 Greifswald, Germany.
| | - Philipp Schick
- University of Greifswald, Institute of Pharmacy, Center of Drug Absorption and Transport (C_DAT), Felix-Hausdorff-Str. 3, D-17489 Greifswald, Germany.
| | - Werner Weitschies
- University of Greifswald, Institute of Pharmacy, Center of Drug Absorption and Transport (C_DAT), Felix-Hausdorff-Str. 3, D-17489 Greifswald, Germany.
| |
Collapse
|
6
|
Blagih J, Hennequart M, Zani F. Tissue Nutrient Environments and Their Effect on Regulatory T Cell Biology. Front Immunol 2021; 12:637960. [PMID: 33868263 PMCID: PMC8050341 DOI: 10.3389/fimmu.2021.637960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Regulatory T cells (Tregs) are essential for mitigating inflammation. Tregs are found in nearly every tissue and play either beneficial or harmful roles in the host. The availability of various nutrients can either enhance or impair Treg function. Mitochondrial oxidative metabolism plays a major role in supporting Treg differentiation and fitness. While Tregs rely heavily on oxidation of fatty acids to support mitochondrial activity, they have found ways to adapt to different tissue types, such as tumors, to survive in competitive environments. In addition, metabolic by-products from commensal organisms in the gut also have a profound impact on Treg differentiation. In this review, we will focus on the core metabolic pathways engaged in Tregs, especially in the context of tissue nutrient environments, and how they can affect Treg function, stability and differentiation.
Collapse
Affiliation(s)
| | | | - Fabio Zani
- The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
7
|
Kayisoglu Ö, Schlegel N, Bartfeld S. Gastrointestinal epithelial innate immunity-regionalization and organoids as new model. J Mol Med (Berl) 2021; 99:517-530. [PMID: 33538854 PMCID: PMC8026474 DOI: 10.1007/s00109-021-02043-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/18/2020] [Accepted: 01/19/2021] [Indexed: 12/27/2022]
Abstract
The human gastrointestinal tract is in constant contact with microbial stimuli. Its barriers have to ensure co-existence with the commensal bacteria, while enabling surveillance of intruding pathogens. At the centre of the interaction lies the epithelial layer, which marks the boundaries of the body. It is equipped with a multitude of different innate immune sensors, such as Toll-like receptors, to mount inflammatory responses to microbes. Dysfunction of this intricate system results in inflammation-associated pathologies, such as inflammatory bowel disease. However, the complexity of the cellular interactions, their molecular basis and their development remains poorly understood. In recent years, stem cell-derived organoids have gained increasing attention as promising models for both development and a broad range of pathologies, including infectious diseases. In addition, organoids enable the study of epithelial innate immunity in vitro. In this review, we focus on the gastrointestinal epithelial barrier and its regional organization to discuss innate immune sensing and development.
Collapse
Affiliation(s)
- Özge Kayisoglu
- Research Centre for Infectious Diseases, Institute for Molecular Infection Biology, Julius Maximilians University of Wuerzburg, Wuerzburg, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, Oberduerrbacher Strasse 6, Wuerzburg, Germany
| | - Sina Bartfeld
- Research Centre for Infectious Diseases, Institute for Molecular Infection Biology, Julius Maximilians University of Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
8
|
Wu Q, Li Q, Zhang X, Ntim M, Wu X, Li M, Wang L, Zhao J, Li S. Treatment with Bifidobacteria can suppress Aβ accumulation and neuroinflammation in APP/PS1 mice. PeerJ 2020; 8:e10262. [PMID: 33194428 PMCID: PMC7602682 DOI: 10.7717/peerj.10262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022] Open
Abstract
Background Alzheimer’s disease (AD), being a complex disorder, is affected either by genetic or environmental factors or both. It is observed that there is an excessive accumulation of amyloid β (Aβ) in the extracellular space of the brain. AD is the first neurodegenerative disease in the elderly, and so far there is no effective treatment. In recent years, many studies have reported that Alzheimer’s disease has a relationship with gut microflora, indicating that regulating gut microbiota could offer therapeutic intervention for AD. This study explored the effect Bifidobacteria has in averting AD. Methods WT and APP/PS1 mice were used for the experiments. The mice were randomly assigned to four groups: WT group, WT + Bi group, AD group (APP/PS1 mouse) and AD + Bi group (Bifidobacteria-treated APP/PS1 mouse). Treatment with Bifidobacteria lasted for 6 months and mice were prepared for immunohistochemistry, immunofluorescence, Thioflavin S staining, Western blotting, PCR and Elisa quantitative assay. Results The results show that after 6 months of treatment with Bifidobacteria signiis to be lesficantly reduces Aβ deposition in cortex and hippocampus of AD mice. The level of insoluble Aβ in the hippocampus and cortex of AD+Bi mice was decreased compared with AD mice. Meanwhile, a significant decrease in the level of soluble Aβ in the cortex of AD+Bi mice but not in the hippocampus was observed. The activation of microglia and the release of inflammatory factors were also determined in this study. From the results, Bifidobacteria inhibited microglial activation and reduced IL-1β, TNF-α, IL-4, IL-6 and INF-γ release. Altogether, these results implied that Bifidobacteria can alleviate the pathological changes of AD through various effects.
Collapse
Affiliation(s)
- Qiong Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| | - Qifa Li
- Functional Laboratory, Dalian Medical University, Dalian, China
| | - Xuan Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Michael Ntim
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| | - Xuefei Wu
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| | - Ming Li
- Department of Microecology, Dalian Medical University, Dalian, China
| | - Li Wang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases in Department of Physiology, Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Steinke I, Ghanei N, Govindarajulu M, Yoo S, Zhong J, Amin RH. Drug Discovery and Development of Novel Therapeutics for Inhibiting TMAO in Models of Atherosclerosis and Diabetes. Front Physiol 2020; 11:567899. [PMID: 33192565 PMCID: PMC7658318 DOI: 10.3389/fphys.2020.567899] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus exists as a comorbidity with congestive heart failure (CHF). However, the exact molecular signaling mechanism linking CHF as the major form of mortality from diabetes remains unknown. Type 2 diabetic patients display abnormally high levels of metabolic products associated with gut dysbiosis. One such metabolite, trimethylamine N-oxide (TMAO), has been observed to be directly related with increased incidence of cardiovascular diseases (CVD) in human patients. TMAO a gut-liver metabolite, comes from the metabolic degenerative product trimethylamine (TMA) that is produced from gut microbial metabolism. Elevated levels of TMAO in diabetics and obese patients are observed to have a direct correlation with increased risk for major adverse cardiovascular events. The pro-atherogenic effect of TMAO is attributed to enhancing inflammatory pathways with cholesterol and bile acid dysregulation, promoting foam cell formation. Recent studies have revealed several potential therapeutic strategies for reducing TMAO levels and will be the central focus for the current review. However, few have focused on developing rational drug therapeutics and may be due to the gaps in knowledge for understanding the mechanism by which microbial TMA producing enzymes and hepatic flavin-containing monoxygenase (FMO) can work together in preventing elevation of TMAO levels. Therefore, it is critical to understand the advantages of developing a novel rational drug design strategy that manipulates FMO production of TMAO and TMA production by microbial enzymes. This review will focus on the inspection of FMO manipulation, as well as gut microbiota dysbiosis and its influence on metabolic disorders including cardiovascular disease and describe novel potential pharmacological therapeutic development.
Collapse
Affiliation(s)
- Ian Steinke
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Nila Ghanei
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Manoj Govindarajulu
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| | - Sieun Yoo
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, United States
| | - Juming Zhong
- Department of Anatomy, Physiology and Pharmacology, Auburn University, Auburn, AL, United States
| | - Rajesh H Amin
- Drug Discovery and Development, Auburn University, Auburn, AL, United States
| |
Collapse
|
10
|
Ciobanu L, Tefas C, Oancea DM, Berce C, Vodnar D, Mester A, Onica S, Toma C, Taulescu M. Effect of Lactobacillus plantarum ACTT 8014 on 5-fluorouracil induced intestinal mucositis in Wistar rats. Exp Ther Med 2020; 20:209. [PMID: 33149773 PMCID: PMC7604756 DOI: 10.3892/etm.2020.9339] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 09/29/2020] [Indexed: 12/21/2022] Open
Abstract
Some previous studies reported that probiotics might decrease the severity of chemotherapy-induced mucositis. This study assessed the potential protective effect of Lactobacillus plantarum ATCC 8014 on 5-fluorouracil (5-FU) induced intestinal mucositis in the Wistar rats. The Crl:WI rats were divided into two groups of six animals (F, L) and one group of 5 animals (M). Group L received for 9 days 3.32x109 CFU/ml of Lactobacillus plantarum orally. In the 7th day of the experiment 400 mg of 5-FU was administered intraperitoneally in groups L and F. Group M received only the vehicles. All animals were sacrificed in the 9th day. Eleven histological characteristics of mucositis were quantified from 0 (normal) to 3 (severe) for duodenum, jejunum and colon. Semiquantitative grades measured Toll-like receptor 4 (TLR4) immunopositive cells. The independent groups were analyzed using the Kruskal-Wallis test, Mann-Whitney U test, with a Bonferroni correction for alpha (P≤0.016). In the group F, treated with 5-FU, the most affected areas were the jejunum and the duodenum. The medium score of histological lesions was 27 for jejunum (minimum 25, maximum 32) and 21 for duodenum (minimum 18, maximum 29). Graded microscopic mucosal changes of the jejunum were significantly lower in group L compared with group F (U=0, P=0.009, Mann-Whitney test). The histological changes depicted on the duodenal and colonic mucosa were less severe in group L than in group F, but without reaching the statistical significance (duodenum: U=6, P=0.172, Mann-Whitney test; colon: U=12, P=0.916, Mann-Whitney test). Although the TLR4 immunoexpression was more intense in group L, no significant statistical difference was revealed at duodenum, jejunum or colonic mucosa. Significantly fewer microscopic changes were depicted in L group on the jejunum, suggesting a potential beneficial effect of Lactobacillus plantarum at this level in 5-FU induced mucositis.
Collapse
Affiliation(s)
- Lidia Ciobanu
- Department of Internal Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania.,Department of Gastroenterology, 'Professor Doctor Octavian Fodor' Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Cristian Tefas
- Department of Internal Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania.,Department of Gastroenterology, 'Professor Doctor Octavian Fodor' Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Diana Maria Oancea
- Department of Internal Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Cristian Berce
- Department of Internal Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Dan Vodnar
- Institute of Life Sciences, Faculty of Food Science and Technology, University ofAgricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Alexandru Mester
- Department of Internal Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Sorina Onica
- Department of Internal Medicine, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Corina Toma
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University ofAgricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Marian Taulescu
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University ofAgricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| |
Collapse
|
11
|
Gupta N, Kainthola A, Tiwari M, Agrawala PK. Gut microbiota response to ionizing radiation and its modulation by HDAC inhibitor TSA. Int J Radiat Biol 2020; 96:1560-1570. [PMID: 33001776 DOI: 10.1080/09553002.2020.1830317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
AIM Trichostatin A (TSA) has been shown to mitigate whole body γ-radiation-induced morbidity and mortality. The current study aimed at studying the effects of TSA post-irradiation treatment on gut-microbiota, especially the translocation of the microbes from the intestine to other organs in C57 Bl/6 mice model. MATERIALS AND METHODS On 1st, 3rd 5th 7th 9th 12th and 14th days after various treatments bacteria were isolated from the intestine and nearby organs (mesenteric lymph node, spleen and liver) for further analysis. The jejunum part of all animals was processed for histological analysis. RESULTS The group radiation + drug showed reduced susceptibility to radiation injury as well as microbiota related anomalies compared to the irradiated alone group. This was described by increased microflora in different parts of the GI tract in the radiation + drug group compared to the irradiated group and reduced histopathological damages in the jejunum. Also, a reduced percentage of translocated bacteria were found in different organs of radiation + drug group animals. CONCLUSION TSA treatment post-irradiation could effectively control bacterial translocation as well as GI injury in mice.
Collapse
Affiliation(s)
- Noopur Gupta
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.,Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Anup Kainthola
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Manisha Tiwari
- Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Paban K Agrawala
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
12
|
New Insights of Oral Colonic Drug Delivery Systems for Inflammatory Bowel Disease Therapy. Int J Mol Sci 2020; 21:ijms21186502. [PMID: 32899548 PMCID: PMC7555849 DOI: 10.3390/ijms21186502] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/25/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
Colonic Drug Delivery Systems (CDDS) are especially advantageous for local treatment of inflammatory bowel diseases (IBD). Site-targeted drug release allows to obtain a high drug concentration in injured tissues and less systemic adverse effects, as consequence of less/null drug absorption in small intestine. This review focused on the reported contributions in the last four years to improve the effectiveness of treatments of inflammatory bowel diseases. The work concludes that there has been an increase in the development of CDDS in which pH, specific enzymes, reactive oxygen species (ROS), or a combination of all of these triggers the release. These delivery systems demonstrated a therapeutic improvement with fewer adverse effects. Future perspectives to the treatment of this disease include the elucidation of molecular basis of IBD diseases in order to design more specific treatments, and the performance of more in vivo assays to validate the specificity and stability of the obtained systems.
Collapse
|
13
|
Zhang K, Sun D, Duan C, Chen H, Din AU, Kong X, Qin X, Zhang B. Application of a Faecalibacterium 16S rDNA genetic marker for species identification of dog fecal waste. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:30615-30624. [PMID: 32472511 DOI: 10.1007/s11356-020-09369-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 05/19/2020] [Indexed: 06/11/2023]
Abstract
A dog-associated 16S rDNA genetic marker (ED-1) was designed to detect dog fecal contamination in water through a comparative bioinformatics analysis of Faecalibacterium sequences. For the dog fecal samples, ED-1 had 100% specificity, a high positive rate (89% in dog feces and 92.3% in dog fecal-contaminated water samples), and a low detection limit (107 copies/100 mL) in dog-contaminated water samples. Detection of water samples from seven provinces or cities of China showed that ED-1 was stable enough to be applied in practice. Furthermore, the abundance and diversity of dog gut microbiota from two private house pets (PHP) and Third Military Medical University (TMMU) dogs were estimated by using operational taxonomic units, and the significant differences of dog feces were found, as the PHP dogs have a more diverse diet and closer contact with human than dogs in TMMU. However, ED-1 could detect the feces from the two regions, indicating that ED-1 has good reliability.
Collapse
Affiliation(s)
- Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Da Sun
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325000, China.
| | - Chuanren Duan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| | - Hang Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Ahmad Ud Din
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Xiangjun Kong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, 999078, China
| | - Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Baoyun Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510000, China
| |
Collapse
|
14
|
Fortin O, Aguilar-Uscanga BR, Vu KD, Salmieri S, Lacroix M. Effect of Saccharomyces Boulardii Cell Wall Extracts on Colon Cancer Prevention in Male F344 Rats Treated with 1,2-Dimethylhydrazine. Nutr Cancer 2018; 70:632-642. [DOI: 10.1080/01635581.2018.1460672] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Olivier Fortin
- INRS-Institut Armand-Frappier, Research Laboratories in Sciences Applied to Food, Laval, Quebec, Canada
| | - Blanca R. Aguilar-Uscanga
- Laboratorio de Microbiología Industrial, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara (UdG), Jalisco, Mexico
| | - Khanh D. Vu
- INRS-Institut Armand-Frappier, Research Laboratories in Sciences Applied to Food, Laval, Quebec, Canada
| | - Stephane Salmieri
- INRS-Institut Armand-Frappier, Research Laboratories in Sciences Applied to Food, Laval, Quebec, Canada
| | - Monique Lacroix
- INRS-Institut Armand-Frappier, Research Laboratories in Sciences Applied to Food, Laval, Quebec, Canada
| |
Collapse
|
15
|
Revaiah PC, Kochhar R, Rana SV, Berry N, Ashat M, Dhaka N, Rami Reddy Y, Sinha SK. Risk of small intestinal bacterial overgrowth in patients receiving proton pump inhibitors versus proton pump inhibitors plus prokinetics. JGH OPEN 2018; 2:47-53. [PMID: 30483563 PMCID: PMC6206996 DOI: 10.1002/jgh3.12045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/08/2018] [Accepted: 01/31/2018] [Indexed: 12/17/2022]
Abstract
Background and Aim Intestinal dysmotility is considered a risk factor for small intestinal bacterial overgrowth (SIBO). Prokinetics improve intestinal motility and are often prescribed with proton pump inhibitors (PPIs) in patients with gastroesophageal reflux disease (GERD) and/or functional dyspepsia. The present study aimed to evaluate the prevalence of SIBO and the orocecal transit time (OCTT) in patients taking PPI compared with those taking PPI plus prokinetics. Methods The study is a single‐center, cross‐sectional study. Enrolled patients (with age > 12 years) were divided into two groups: patients taking PPIs for more than 3 months (Group A) and those taking PPIs with prokinetics for more than 3 months (Group B) for various indications. Lactulose breath test (LBT) for OCTT and glucose breath test (GBT) for SIBO were conducted for all patients. Results Of the 147 enrolled patients, SIBO was documented in 13.2% patients in Group A versus 1.8% in Group B, P = 0.018. Median OCTT in Group A was 130 (105–160) min compared with 120 (92.5–147.5) min in Group B (P = 0.010). Median OCTT among SIBO‐positive patients was 160 (140–172.5) min compared with SIBO‐negative patients, where it was 120 (103.75–150) min (P = 0.002). The duration and type of PPI used were not associated with the occurrence of SIBO in our study. Conclusion The use of prokinetics in patients on PPI may reduce the risk of SIBO by enhancing intestinal motility and may reduce SIBO risk associated with long‐term PPI use.
Collapse
Affiliation(s)
- Pruthvi C Revaiah
- Department of Medicine Postgraduate Institute of Medical Education and Research Chandigarh India
| | - Rakesh Kochhar
- Department of Gastroenterology Postgraduate Institute of Medical Education and Research Chandigarh India
| | - Surinder V Rana
- Department of Gastroenterology Postgraduate Institute of Medical Education and Research Chandigarh India
| | - Neha Berry
- Department of Gastroenterology Postgraduate Institute of Medical Education and Research Chandigarh India
| | - Munish Ashat
- Department of Gastroenterology Postgraduate Institute of Medical Education and Research Chandigarh India
| | - Narendra Dhaka
- Department of Gastroenterology Postgraduate Institute of Medical Education and Research Chandigarh India
| | - Y Rami Reddy
- Department of Gastroenterology Postgraduate Institute of Medical Education and Research Chandigarh India
| | - Saroj K Sinha
- Department of Gastroenterology Postgraduate Institute of Medical Education and Research Chandigarh India
| |
Collapse
|
16
|
Mancuso C, Santangelo R. Alzheimer's disease and gut microbiota modifications: The long way between preclinical studies and clinical evidence. Pharmacol Res 2017; 129:329-336. [PMID: 29233677 DOI: 10.1016/j.phrs.2017.12.009] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/15/2022]
Abstract
Recent studies have suggested the role of an infectious component in the pathogenesis of Alzheimer's disease (AD). In light of this, research has focused on some bacteria constituting the intestinal microbial flora which can produce amyloid. Once generated, the latter hypothetically triggers a systemic inflammatory response which compromises complex brain functions, such as learning and memory. Clinical studies have shown that, in cognitively impaired elderly patients with brain amyloidosis, there is lower abundance in the gut of E. rectale and B. fragilis, two bacterial species which have an anti-inflammatory activity, versus a greater amount of pro-inflammatory genera such as Escherichia/Shigella. According to these findings, some clinical studies have demonstrated that supplementation with Lactobacilli- and Bifidobacteria- based probiotics has improved cognitive, sensory and emotional functions in subjects with AD. Moreover, certain herbal products, in particular dietetic polyphenols, have proved capable of restoring dysbiosis and, therefore, their prebiotic role could be effective in counteracting the onset of AD regardless of their activity of free radical scavenging or enhancement of the cell stress response. One of the recent greatest novelties in the field of neurodegenerative diseases is the chance to prevent or slow down AD progression with agents, such as probiotics and prebiotics, acting outside the central nervous system.
Collapse
Affiliation(s)
- Cesare Mancuso
- Institute of Pharmacology, Catholic University School of Medicine, Largo F. Vito, 1-00168 Rome, Italy.
| | - Rosaria Santangelo
- Institute of Microbiology, Catholic University School of Medicine, Largo F. Vito, 1-00168 Rome, Italy
| |
Collapse
|
17
|
Bacterial Chat: Intestinal Metabolites and Signals in Host-Microbiota-Pathogen Interactions. Infect Immun 2017; 85:IAI.00476-17. [PMID: 28947641 DOI: 10.1128/iai.00476-17] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Intestinal bacteria employ microbial metabolites from the microbiota and chemical signaling during cell-to-cell communication to regulate several cellular functions. Pathogenic bacteria are extremely efficient in orchestrating their response to these signals through complex signaling transduction systems. Precise coordination and interpretation of these multiple chemical cues is important within the gastrointestinal (GI) tract. Enteric foodborne pathogens, such as enterohemorrhagic Escherichia coli (EHEC) and Salmonella enterica serovar Typhimurium, or the surrogate murine infection model for EHEC, Citrobacter rodentium, are all examples of microorganisms that modulate the expression of their virulence repertoire in response to signals from the microbiota or the host, such as autoinducer-3 (AI-3), epinephrine (Epi), and norepinephrine (NE). The QseBC and QseEF two-component systems, shared by these pathogens, are involved in sensing these signals. We review how these signaling systems sense and relay these signals to drive bacterial gene expression; specifically, to modulate virulence. We also review how bacteria chat via chemical signals integrated with metabolite recognition and utilization to promote successful associations among enteric pathogens, the microbiota, and the host.
Collapse
|
18
|
Analysis of the Duodenal Microbiome in Autistic Individuals: Association With Carbohydrate Digestion. J Pediatr Gastroenterol Nutr 2017; 64:e110-e116. [PMID: 27811623 DOI: 10.1097/mpg.0000000000001458] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES There is evidence that symptoms of maldigestion or malabsorption in autistic individuals are related to changes in the indigenous microbiota. Analysis of colonic bacteria has revealed microbial dysbiosis in children with autism; however, characteristics of the duodenal microbiome are not well described. In the present study the microbiome of the duodenal mucosa of subjects with autism was evaluated for dysbiosis, bacteria overgrowth, and microbiota associated with carbohydrate digestion. The relationship between the duodenal microbiome and disaccharidase activity was analyzed in biopsies from 21 autistic subjects and 19 children without autism. METHODS Microbiota composition was determined by 16S ribosomal RNA gene sequencing, and disaccharidase activity via biochemical assays. RESULTS Although subjects with autism had a higher frequency of constipation (P < 0.005), there was no difference in disaccharidase activity between groups. In addition, no differences in microbiome diversity (species richness and evenness) were observed. Bacteria belonging to the genus Burkholderia were more abundant in subjects with autism, whereas members of the genus Neisseria were less abundant. At the species level, a relative decrease in abundance of 2 Bacteroides species and Escherichia coli was found in autistic individuals. There was a positive correlation between the abundance of Clostridium species, and disaccharidase activity, in autistic individuals. CONCLUSIONS There are a variety of changes at the genus and species level in duodenal microbiota in children with autism that could be influenced by carbohydrate malabsorption. These observations could be affected by variations in individual diets, but also may represent a more pervasive dysbiosis that results in metabolites that affect the behavior of autistic children.
Collapse
|
19
|
Dong WW, Xuan FL, Zhong FL, Jiang J, Wu S, Li D, Quan LH. Comparative Analysis of the Rats' Gut Microbiota Composition in Animals with Different Ginsenosides Metabolizing Activity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:327-337. [PMID: 28025886 DOI: 10.1021/acs.jafc.6b04848] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Following oral intake of Panax ginseng, major ginsenosides are metabolized to deglycosylated ginsenosides by gut microbiota before absorption into the blood. As the composition of gut microbiota varies between individuals, metabolic activities are significantly different. We selected 6 rats with low efficiency metabolism (LEM) and 6 rats with high efficiency metabolism (HEM) from 60 rats following oral administration of Panax ginseng extract, and analyzed their gut microbiota composition using Illumina HiSeq sequencing of the 16S rRNA gene. The components of gut microbiota between the LEM and HEM groups were significantly different. Between the 2 groups, S24-7, Alcaligenaceae, and Erysipelotrichaceae occupied most OTUs of the HEM group, which was notably higher than the LEM group. Furthermore, we isolated Bifidobacterium animalis GM1 that could convert the ginsenoside Rb1 to Rd. The result implies that these specific intestinal bacteria may dominate the metabolism of Panax ginseng.
Collapse
Affiliation(s)
- Wei-Wei Dong
- Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular ( Yanbian University ), Ministry of Education, Park Road 977, Yanji City, Jilin Province 133002, China
| | - Fang-Ling Xuan
- Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular ( Yanbian University ), Ministry of Education, Park Road 977, Yanji City, Jilin Province 133002, China
| | - Fei-Liang Zhong
- Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular ( Yanbian University ), Ministry of Education, Park Road 977, Yanji City, Jilin Province 133002, China
| | - Jun Jiang
- Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular ( Yanbian University ), Ministry of Education, Park Road 977, Yanji City, Jilin Province 133002, China
| | - Songquan Wu
- Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular ( Yanbian University ), Ministry of Education, Park Road 977, Yanji City, Jilin Province 133002, China
| | - Donghao Li
- Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular ( Yanbian University ), Ministry of Education, Park Road 977, Yanji City, Jilin Province 133002, China
| | - Lin-Hu Quan
- Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular ( Yanbian University ), Ministry of Education, Park Road 977, Yanji City, Jilin Province 133002, China
| |
Collapse
|
20
|
Dong WW, Zhao J, Zhong FL, Zhu WJ, Jiang J, Wu S, Yang DC, Li D, Quan LH. Biotransformation of Panax ginseng extract by rat intestinal microflora: identification and quantification of metabolites using liquid chromatography-tandem mass spectrometry. J Ginseng Res 2017; 41:540-547. [PMID: 29021702 PMCID: PMC5628354 DOI: 10.1016/j.jgr.2016.11.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 10/06/2016] [Accepted: 11/21/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In general, after Panax ginseng is administered orally, intestinal microbes play a crucial role in its degradation and metabolization process. Studies on the metabolism of P. ginseng by microflora are important for obtaining a better understanding of their biological effects. METHODS In vitro biotransformation of P. ginseng extract by rat intestinal microflora was investigated at 37°C for 24 h, and the simultaneous determination of the metabolites and metabolic profile of P. ginseng saponins by rat intestinal microflora was achieved using LC-MS/MS. RESULTS A total of seven ginsenosides were detected in the P. ginseng extract, including ginsenosides Rg1, Re, Rf, Rb1, Rc, Rb2, and Rd. In the transformed P. ginseng samples, considerable amounts of deglycosylated metabolite compound K and Rh1 were detected. In addition, minimal amounts of deglycosylated metabolites (ginsenosides Rg2, F1, F2, Rg3, and protopanaxatriol-type ginsenosides) and untransformed ginsenosides Re, Rg1, and Rd were detected at 24 h. The results indicated that the primary metabolites are compound K and Rh1, and the protopanaxadiol-type ginsenosides were more easily metabolized than protopanaxatriol-type ginsenosides. CONCLUSION This is the first report of the identification and quantification of the metabolism and metabolic profile of P. ginseng extract in rat intestinal microflora using LC-MS/MS. The current study provided new insights for studying the metabolism and active metabolites of P. ginseng.
Collapse
Affiliation(s)
- Wei-Wei Dong
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| | - Jinhua Zhao
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| | - Fei-Liang Zhong
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| | - Wen-Jing Zhu
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| | - Jun Jiang
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| | - Songquan Wu
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| | - Deok-Chun Yang
- Department of Oriental Medicinal Material and Processing, College of Life Science, Korean Ginseng Center Most Valuable Product and Ginseng Genetic Resource Bank, Kyung Hee University, Yongin, Republic of Korea
| | - Donghao Li
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| | - Lin-Hu Quan
- Key Laboratory of Natural Resources of Changbai Mountain and Functional Molecules (Yanbian University), Ministry of Education, Yangji, China
| |
Collapse
|
21
|
Tas S, Ozkul F, Arik MK, Kiraz A, Vural A. The effect of amifostine on bacterial translocation after radiation ınduced acute enteritis. Acta Cir Bras 2016; 31:156-60. [DOI: 10.1590/s0102-865020160030000002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/15/2016] [Indexed: 11/22/2022] Open
Affiliation(s)
- Sukru Tas
- Çanakkale Onsekiz Mart University, Turkey
| | | | | | - Asli Kiraz
- Çanakkale Onsekiz Mart University, Turkey
| | | |
Collapse
|
22
|
Vaikunthanathan T, Safinia N, Lombardi G, Lechler RI. Microbiota, immunity and the liver. Immunol Lett 2016; 171:36-49. [PMID: 26835593 DOI: 10.1016/j.imlet.2016.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/24/2016] [Accepted: 01/27/2016] [Indexed: 12/18/2022]
Abstract
The gut harbors a complex community of over 100 trillion microbial cells known to exist in symbiotic harmony with the host influencing human physiology, metabolism, nutrition and immune function. It is now widely accepted that perturbations of this close partnership results in the pathogenesis of several major diseases with increasing evidence highlighting their role outside of the intestinal tract. The intimate proximity and circulatory loop of the liver and the gut has attracted significant attention regarding the role of the microbiota in the development and progression of liver disease. Here we give an overview of the interaction between the microbiota and the immune system and focus on their convincing role in both the propagation and treatment of liver disease.
Collapse
Affiliation(s)
- T Vaikunthanathan
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| | - N Safinia
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| | - G Lombardi
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| | - R I Lechler
- MRC Centre for Transplantation, Division of Transplantation Immunology & Mucosal Biology, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.
| |
Collapse
|
23
|
Mann ER, Bernardo D, English NR, Landy J, Al-Hassi HO, Peake STC, Man R, Elliott TR, Spranger H, Lee GH, Parian A, Brant SR, Lazarev M, Hart AL, Li X, Knight SC. Compartment-specific immunity in the human gut: properties and functions of dendritic cells in the colon versus the ileum. Gut 2016; 65:256-70. [PMID: 25666191 PMCID: PMC4530083 DOI: 10.1136/gutjnl-2014-307916] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 11/27/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Dendritic cells (DC) mediate intestinal immune tolerance. Despite striking differences between the colon and the ileum both in function and bacterial load, few studies distinguish between properties of immune cells in these compartments. Furthermore, information of gut DC in humans is scarce. We aimed to characterise human colonic versus ileal DC. DESIGN Human DC from paired colonic and ileal samples were characterised by flow cytometry, electron microscopy or used to stimulate T cell responses in a mixed leucocyte reaction. RESULTS A lower proportion of colonic DC produced pro-inflammatory cytokines (tumour necrosis factor-α and interleukin (IL)-1β) compared with their ileal counterparts and exhibited an enhanced ability to generate CD4(+)FoxP3(+)IL-10(+) (regulatory) T cells. There were enhanced proportions of CD103(+)Sirpα(-) DC in the colon, with increased proportions of CD103(+)Sirpα(+) DC in the ileum. A greater proportion of colonic DC subsets analysed expressed the lymph-node-homing marker CCR7, alongside enhanced endocytic capacity, which was most striking in CD103(+)Sirpα(+) DC. Expression of the inhibitory receptor ILT3 was enhanced on colonic DC. Interestingly, endocytic capacity was associated with CD103(+) DC, in particular CD103(+)Sirpα(+) DC. However, expression of ILT3 was associated with CD103(-) DC. Colonic and ileal DC differentially expressed skin-homing marker CCR4 and small-bowel-homing marker CCR9, respectively, and this corresponded to their ability to imprint these homing markers on T cells. CONCLUSIONS The regulatory properties of colonic DC may represent an evolutionary adaptation to the greater bacterial load in the colon. The colon and the ileum should be regarded as separate entities, each comprising DC with distinct roles in mucosal immunity and imprinting.
Collapse
Affiliation(s)
- Elizabeth R Mann
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK,Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David Bernardo
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Nicholas R English
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Jon Landy
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK,St. Mark's Hospital, North West London Hospitals NHS Trust, Harrow, UK
| | - Hafid O Al-Hassi
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Simon TC Peake
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK,St. Mark's Hospital, North West London Hospitals NHS Trust, Harrow, UK
| | - Ripple Man
- St. Mark's Hospital, North West London Hospitals NHS Trust, Harrow, UK
| | - Timothy R Elliott
- St. Mark's Hospital, North West London Hospitals NHS Trust, Harrow, UK
| | - Henning Spranger
- St. Mark's Hospital, North West London Hospitals NHS Trust, Harrow, UK
| | - Gui Han Lee
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Alyssa Parian
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven R Brant
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Lazarev
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ailsa L Hart
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK,St. Mark's Hospital, North West London Hospitals NHS Trust, Harrow, UK
| | - Xuhang Li
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stella C Knight
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| |
Collapse
|
24
|
Microbial deglycosylation and ketonization of ginsenoside by Cladosporium cladosporioide and their anticancer activity. Antonie Van Leeuwenhoek 2015; 109:179-85. [PMID: 26558793 DOI: 10.1007/s10482-015-0619-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 11/05/2015] [Indexed: 10/22/2022]
Abstract
Ginseng has been used for thousands of years in Asian countries as a traditional medicinal herb and has gained great popularity in the past decade. Ginsenosides are the major pharmacological components in ginseng. We here show that Cladosporium cladosporioide is able to convert the major ginsenoside Rb1 into four known metabolites (ginsenosides Rd, F2, CK and PPD) and two new metabolites [12β-hydroxydammar-3-one-20(S)-O-β-D-glucopyranoside (3-oxo-CK) and dammar-24-en-12β,20(S)-diol-3-one (3-oxo-PPD)]. CK, PPD and 3-oxo-PPD were shown to have a potent antiproliferative activity against A549 lung cancer cells. We found that Rb1 → Rd → F2 → CK → PPD or 3-oxo-CK → 3-oxo-PPD represents the ginsenoside metabolic pathway.
Collapse
|
25
|
Barboza JL, Okun MS, Moshiree B. The treatment of gastroparesis, constipation and small intestinal bacterial overgrowth syndrome in patients with Parkinson's disease. Expert Opin Pharmacother 2015; 16:2449-64. [PMID: 26374094 DOI: 10.1517/14656566.2015.1086747] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) affects the nerves of the entire gastrointestinal (GI) tract and may result in profound gastrointestinal (GI) dysfunction leading to poor patient outcomes. Common GI disturbances in patients with PD include gastroparesis (GP), constipation and small intestinal bacterial overgrowth syndrome (SIBO). In particular, GP is difficult to treat due to the limited options available and precautions, contraindications and adverse effects associated with the approved treatments. Moreover, some commonly used medications can worsen pre-existing PD. AREAS COVERED Our review will focus on treatment options for GP and SIBO with motilin agonists, dopamine receptor antagonists, Ghrelin agonists muscarinic agonists, 5-HT4 receptor agonists, antibiotics, probiotics and herbal formulation such as iberogast. Constipation occurs in the majority of patients with PD and fortunately many treatments are now available. Our review is based on original papers or reviews selected from PUBMED search and Cochrane reviews. EXPERT OPINION Motility disorders of the GI tract are found frequently in patients with PD and treating the underlying GI disorders caused by PD with various prokinetics and laxatives is paramount in achieving improvements in patient's motor function. Various prokinetics and laxatives are now available to provide some relief of the GI morbidity caused by PD leading even to better absorption of even the PD treatments.
Collapse
Affiliation(s)
| | - Michael S Okun
- b 2 University of Florida, Center for Movement Disorders and Neurorestoration , Gainesville, FL, UK
| | | |
Collapse
|
26
|
Corstens MN, Berton-Carabin CC, de Vries R, Troost FJ, Masclee AAM, Schroën K. Food-grade micro-encapsulation systems that may induce satiety via delayed lipolysis: A review. Crit Rev Food Sci Nutr 2015; 57:2218-2244. [DOI: 10.1080/10408398.2015.1057634] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Meinou N. Corstens
- Department of Agrotechnology & Food Sciences, Food Process Engineering Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Claire C. Berton-Carabin
- Department of Agrotechnology & Food Sciences, Food Process Engineering Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Renko de Vries
- Department of Agrotechnology & Food Sciences, Physical Chemistry and Colloid Science Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Freddy J. Troost
- Department of Internal Medicine, Division of Gastroenterology-Hepatology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ad A. M. Masclee
- Department of Internal Medicine, Division of Gastroenterology-Hepatology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Karin Schroën
- Department of Agrotechnology & Food Sciences, Food Process Engineering Group, Wageningen University and Research Center, Wageningen, The Netherlands
| |
Collapse
|
27
|
Cancer preventive effects of a specific probiotic fermented milk containing Lactobacillus acidophilus CL1285, L. casei LBC80R and L. rhamnosus CLR2 on male F344 rats treated with 1,2-dimethylhydrazine. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
28
|
Yoo DH, Kim DH. Lactobacillus pentosus var. plantarum C29 increases the protective effect of soybean against scopolamine-induced memory impairment in mice. Int J Food Sci Nutr 2015; 66:912-8. [PMID: 26171634 DOI: 10.3109/09637486.2015.1064865] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 06/02/2015] [Accepted: 06/03/2015] [Indexed: 02/05/2023]
Abstract
Biological activities of soybean saponins are dependent on their metabolism by gut microbiota, which generate absorbable bioactive metabolites. Therefore, to enhance the pharmacological effect of soybean, we fermented defatted soybean powder (SP) with Lactobacillus pentosus var. plantarum C29 and measured its protective effect against scopolamine-induced memory impairment in mice using the passive avoidance, Y-maze and Morris water maze tasks. Fermentation increased soyasapogenol B, genistein and daidzein content of soybean and enhanced the protective effect of soybean against scopolamine-induced memory impairment. Additionally, compared with the exthanol extract of soybean, fermented SP (FSP) increased the expression of brain-derived neurotrophic factor (BDNF) in the hippocampi of scopolamine-treated mice. Furthermore, FSP inhibited acetylcholinesterase (AChE) activity in vitro and ex vivo. These findings suggest that C29 fermentation might increase the ameliorating effect of soybean against memory impairments by inhibiting AChE activity and increasing BDNF expression.
Collapse
Affiliation(s)
- Dae-Hyoung Yoo
- a Department of Life and Nanopharmaceutical Sciences , College of Pharmacy, Kyung Hee University , Seoul , Korea
| | - Dong-Hyun Kim
- a Department of Life and Nanopharmaceutical Sciences , College of Pharmacy, Kyung Hee University , Seoul , Korea
| |
Collapse
|
29
|
Vanhoecke B, Bateman E, Mayo B, Vanlancker E, Stringer A, Thorpe D, Keefe D. Dark Agouti rat model of chemotherapy-induced mucositis: establishment and current state of the art. Exp Biol Med (Maywood) 2015; 240:725-41. [PMID: 25966981 PMCID: PMC4935219 DOI: 10.1177/1535370215581309] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mucositis is a major oncological problem. The entire gastrointestinal and genitourinary tract and also other mucosal surfaces can be affected in recipients of radiotherapy, and/or chemotherapy. Major progress has been made in recent years in understanding the mechanisms of oral and small intestinal mucositis, which appears to be more prominent than colonic damage. This progress is largely due to the development of representative laboratory animal models of mucositis. This review focuses on the development and establishment of the Dark Agouti rat mammary adenocarcinoma model by the Mucositis Research Group of the University of Adelaide over the past 20 years to characterize the mechanisms underlying methotrexate-, 5-fluorouracil-, and irinotecan-induced mucositis. It also aims to summarize the results from studies using different animal model systems to identify new molecular and cellular markers of mucositis.
Collapse
Affiliation(s)
- Barbara Vanhoecke
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia Laboratory of Microbial Ecology and Technology, University of Ghent, 9000 Ghent, Belgium
| | - Emma Bateman
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia
| | - Bronwen Mayo
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia Sansom Institute for Health Research, University of South Australia, Adelaide, 5001 South Australia, Australia
| | - Eline Vanlancker
- Laboratory of Microbial Ecology and Technology, University of Ghent, 9000 Ghent, Belgium
| | - Andrea Stringer
- Sansom Institute for Health Research, University of South Australia, Adelaide, 5001 South Australia, Australia
| | - Daniel Thorpe
- Sansom Institute for Health Research, University of South Australia, Adelaide, 5001 South Australia, Australia
| | - Dorothy Keefe
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia Director, SA Cancer Service, Royal Adelaide Hospital, Adelaide, 5005 South Australia, Australia
| |
Collapse
|
30
|
Dalhoff AA, Levy SB. Does use of the polyene natamycin as a food preservative jeopardise the clinical efficacy of amphotericin B? A word of concern. Int J Antimicrob Agents 2015; 45:564-7. [DOI: 10.1016/j.ijantimicag.2015.02.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 11/28/2022]
|
31
|
Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev 2014; 38:996-1047. [PMID: 24861948 PMCID: PMC4262072 DOI: 10.1111/1574-6976.12075] [Citation(s) in RCA: 774] [Impact Index Per Article: 70.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/29/2014] [Accepted: 05/09/2014] [Indexed: 02/06/2023] Open
Abstract
The microorganisms that inhabit the human gastrointestinal tract comprise a complex ecosystem with functions that significantly contribute to our systemic metabolism and have an impact on health and disease. In line with its importance, the human gastrointestinal microbiota has been extensively studied. Despite the fact that a significant part of the intestinal microorganisms has not yet been cultured, presently over 1000 different microbial species that can reside in the human gastrointestinal tract have been identified. This review provides a systematic overview and detailed references of the total of 1057 intestinal species of Eukarya (92), Archaea (8) and Bacteria (957), based on the phylogenetic framework of their small subunit ribosomal RNA gene sequences. Moreover, it unifies knowledge about the prevalence, abundance, stability, physiology, genetics and the association with human health of these gastrointestinal microorganisms, which is currently scattered over a vast amount of literature published in the last 150 years. This detailed physiological and genetic information is expected to be instrumental in advancing our knowledge of the gastrointestinal microbiota. Moreover, it opens avenues for future comparative and functional metagenomic and other high-throughput approaches that need a systematic and physiological basis to have an impact.
Collapse
Affiliation(s)
- Mirjana Rajilić-Stojanović
- Department for Biotechnology and Biochemical Engineering, Faculty of Technology and Metallurgy, University of BelgradeBelgrade, Serbia
- Laboratory of Microbiology, Wageningen UniversityWageningen, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen UniversityWageningen, The Netherlands
- Departments of Bacteriology and Immunology, and Veterinary Biosciences, University of HelsinkiHelsinki, Finland
| |
Collapse
|
32
|
Enhanced viability of probiotic Saccharomyces boulardii encapsulated by layer-by-layer approach in pH responsive chitosan–dextran sulfate polyelectrolytes. J FOOD ENG 2014. [DOI: 10.1016/j.jfoodeng.2014.03.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
33
|
Dubourg G, Lagier JC, Robert C, Armougom F, Hugon P, Metidji S, Dione N, Dangui NPM, Pfleiderer A, Abrahao J, Musso D, Papazian L, Brouqui P, Bibi F, Yasir M, Vialettes B, Raoult D. Culturomics and pyrosequencing evidence of the reduction in gut microbiota diversity in patients with broad-spectrum antibiotics. Int J Antimicrob Agents 2014; 44:117-24. [DOI: 10.1016/j.ijantimicag.2014.04.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/25/2014] [Accepted: 04/25/2014] [Indexed: 12/20/2022]
|
34
|
Rashid MU, Dalhoff A, Bäckström T, Björkhem-Bergman L, Panagiotidis G, Weintraub A, Nord CE. Ecological impact of MCB3837 on the normal human microbiota. Int J Antimicrob Agents 2014; 44:125-30. [PMID: 24931053 DOI: 10.1016/j.ijantimicag.2014.03.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/26/2014] [Accepted: 03/30/2014] [Indexed: 11/30/2022]
Abstract
MCB3837 is a novel, water-soluble, injectable prodrug that is rapidly converted to the active substance MCB3681 in vivo following intravenous (i.v.) administration. Both MCB3837 and MCB3681 are oxazolidinone-quinolone hybrid molecules. The purpose of the present study was to investigate the effect of MCB3681 on the human skin, nose, oropharyngeal and intestinal microbiota following administration of MCB3837. Twelve healthy male subjects received i.v. MCB3837 (6 mg/kg body weight) once daily for 5 days. Skin, nose, saliva and faecal samples were collected on Day -1 (pre dose), during administration on Days 2 and 5, and post dose on Days 8, 12 and 19. Micro-organisms were identified to genus level. No measurable concentrations of MCB3681 were found in any saliva samples or in the faecal samples on Day -1. On Day 2, 10 volunteers had faecal MCB3681 concentrations between 16.5 mg/kg faeces and 275.1mg/kg faeces; no MCB3681 in faeces could be detected in two of the volunteers. On Day 5, all volunteers had faecal concentrations of MCB3681 ranging from 98.9 to 226.3 mg/kg. MCB3681 caused no ecological changes in the skin, nasal and oropharyngeal microbiota. The numbers of enterococci, bifidobacteria, lactobacilli and clostridia decreased in the intestinal microbiota during administration of the drug. Numbers of Escherichia coli, other enterobacteria and Candida were not affected during the study. There was no impact on the number of Bacteroides. The faecal microbiota was normalised on Day 19. No new colonising aerobic or anaerobic Gram-positive bacteria with MCB3681 minimum inhibitory concentrations of ≥4 mg/L were found.
Collapse
Affiliation(s)
- Mamun-Ur Rashid
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Axel Dalhoff
- University Medical Center Schleswig-Holstein, Institute for Infection Medicine, Brunswiker Str. 4, D-24105 Kiel, Germany
| | - Tobias Bäckström
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Linda Björkhem-Bergman
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Georgios Panagiotidis
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Andrej Weintraub
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Carl Erik Nord
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
35
|
Abstract
The purpose of this essay is threefold: to give an outline of the life and the various achievements of Theodor Escherich, to provide a background to his discovery of what he called Bacterium coli commune (now Escherichia coli), and to indicate the enormous impact of studies with this organism, long before it became the cornerstone of research in bacteriology and in molecular biology.
Collapse
|
36
|
Shafquat A, Joice R, Simmons SL, Huttenhower C. Functional and phylogenetic assembly of microbial communities in the human microbiome. Trends Microbiol 2014; 22:261-6. [PMID: 24618403 DOI: 10.1016/j.tim.2014.01.011] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/29/2014] [Accepted: 01/31/2014] [Indexed: 12/28/2022]
Abstract
Microbial communities associated with the human body, that is, the human microbiome, are complex ecologies critical for normal development and health. The taxonomic and phylogenetic composition of these communities tends to significantly differ among individuals, precluding the definition of a simple, shared set of 'core' microbes. Here, we review recent evidence and ecological theory supporting the assembly of host-associated microbial communities in terms of functional traits rather than specific organisms. That is, distinct microbial species may be responsible for specific host-associated functions and phenotypes in distinct hosts. We discuss how ecological processes (selective and stochastic forces) governing the assembly of metazoan communities can be adapted to describe microbial ecologies in host-associated environments, resulting in both niche-specific and 'core' metabolic and other pathways maintained throughout the human microbiome. The extent to which phylogeny and functional traits are linked in host-associated microbes, as opposed to unlinked by mechanisms, such as lateral transfer, remains to be determined. However, the definition of these functional assembly rules within microbial communities using controlled model systems and integrative 'omics' represents a fruitful opportunity for molecular systems ecology.
Collapse
Affiliation(s)
- Afrah Shafquat
- Department of Biostatistics, Harvard School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA
| | - Regina Joice
- Department of Biostatistics, Harvard School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA
| | - Sheri L Simmons
- Bay Paul Center, Marine Biological Laboratory, 7 MBL Street, Woods Hole, MA 02543, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA; The Broad Institute of Harvard and Massachusetts Institute of Technology (MIT), 301 Binney Street, Cambridge, MA 02142, USA.
| |
Collapse
|
37
|
Vieira NA, Borgo HC, da Silva Dalben G, Bachega MI, Pereira PCM. Evaluation of fecal microorganisms of children with cleft palate before and after palatoplasty. Braz J Microbiol 2014; 44:835-8. [PMID: 24516450 PMCID: PMC3910197 DOI: 10.1590/s1517-83822013000300026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 11/13/2012] [Indexed: 01/12/2023] Open
Abstract
This study isolated and quantified intestinal bacteria of children with cleft palate before and after palatoplasty. A prospective study was conducted from May 2007 to September 2008 on 18 children with cleft palate, aged one to four years, of both genders, attending a tertiary cleft center in Brazil for palatoplasty, to analyze the effect of surgical palate repair on the concentration of anaerobes Bacteroides sp, Bifidobacterium sp and microaerophiles Lactobacillus sp in feces of infants with cleft palate before and 24 hours after treatment with cefazolin for palatoplasty. There was significant reduction of Lactobacillus sp (p < 0.002), Bacteroides sp (p < 0.001) and Bifidobacterium sp (p = 0.021) after palatoplasty, revealing that surgery and utilization of cefazolin significantly influenced the fecal microbiota comparing collections before and after surgery. However, due to study limitations, it was not possible to conclude that other isolated factors, such as surgical stress, anesthetics and other medications used in palatoplasty might have a significant influence on the microbiota. Considering the important participation of the intestinal microbiota on both local and systemic metabolic and immunological activities of the host, professionals should be attentive to the possible influence of these changes in patients submitted to cleft repair.
Collapse
Affiliation(s)
- Narciso Almeida Vieira
- Clinical Analysis Laboratory, Hospital for Rehabilitation of Craniofacial Anomalies, Universidade de São Paulo, Bauru, SP, Brazil
| | - Hilton Coimbra Borgo
- Clinical Analysis Laboratory, Hospital for Rehabilitation of Craniofacial Anomalies, Universidade de São Paulo, Bauru, SP, Brazil
| | - Gisele da Silva Dalben
- Clinical Analysis Laboratory, Hospital for Rehabilitation of Craniofacial Anomalies, Universidade de São Paulo, Bauru, SP, Brazil
| | - Maria Irene Bachega
- Clinical Analysis Laboratory, Hospital for Rehabilitation of Craniofacial Anomalies, Universidade de São Paulo, Bauru, SP, Brazil
| | - Paulo Câmara Marques Pereira
- Tropical Diseases Department, Escola de Medicina, Universidade Estadual de São Paulo "Júlio de Mesquita Filho", Botucatu, SP, Brazil
| |
Collapse
|
38
|
Salim SY, Young PY, Lukowski CM, Madsen KL, Sis B, Churchill TA, Khadaroo RG. VSL#3 probiotics provide protection against acute intestinal ischaemia/reperfusion injury. Benef Microbes 2013; 4:357-365. [PMID: 24240573 DOI: 10.3920/bm2013.0026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Acute intestinal ischaemia/reperfusion injury (AII/R) is an adaptive physiologic response during critical illness, involving mesenteric vasoconstriction and hypoperfusion. Prevention of AII/R in high risk patient populations would have a significant impact on morbidity and mortality. The purpose of this study was to investigate the protective effects of VSL#3 probiotic treatment in a murine model of AII/R. Adult 129/SvEv mice were subjected to an experimental AII/R model using superior mesenteric artery occlusion. Animals were pre-treated with either three days or two weeks of VSL#3 probiotics. Local tissue injury markers were assessed by levels of myeloperoxidase and activation of nuclear factor kappa B (NFкB). Systemic and local cytokines, including interleukin (IL)-1β, IL- 10, TNFα, and interferon gamma were measured by ELISA and multiplex fluorescent detection. VSL#3 probiotics reduced local tissue inflammation and injury due to AII/R. A two-week course of VSL#3 was more effective than a shorter three-day course. The reduction in local inflammation from the two-week course of VSL#3 is correlated to a significant reduction in levels of active IL-1β, and tissue levels of myeloperoxidase. Levels of active NFкB were significantly elevated in the vehicle-fed AII/R mice, corroborating with tissue inflammation, which were attenuated by VSL#3 administrations. VSL#3 did not cause any systemic inflammation or lung injury. VSL#3 probiotics are effective in reducing local tissue injury from AII/R by down-regulating pro-inflammatory mediators and immune cell recruitment. This study highlights a potential role for VSL#3 in management of patients at high risk for AII/R.
Collapse
Affiliation(s)
- S Y Salim
- Department of Surgery, University of Alberta, 2D WMC, 8440-112 St NW, Edmonton, AB T6G 2B7, Canada
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - P Y Young
- Department of Surgery, University of Alberta, 2D WMC, 8440-112 St NW, Edmonton, AB T6G 2B7, Canada
| | - C M Lukowski
- Department of Surgery, University of Alberta, 2D WMC, 8440-112 St NW, Edmonton, AB T6G 2B7, Canada
| | - K L Madsen
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - B Sis
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - T A Churchill
- Department of Surgery, University of Alberta, 2D WMC, 8440-112 St NW, Edmonton, AB T6G 2B7, Canada
| | - R G Khadaroo
- Department of Surgery, University of Alberta, 2D WMC, 8440-112 St NW, Edmonton, AB T6G 2B7, Canada
- Division of Critical Care Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| |
Collapse
|
39
|
Madodé YE, Nout MJ, Bakker EJ, Linnemann AR, Hounhouigan DJ, van Boekel MA. Enhancing the digestibility of cowpea (Vigna unguiculata) by traditional processing and fermentation. Lebensm Wiss Technol 2013. [DOI: 10.1016/j.lwt.2013.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
40
|
Sex related differences on valproic acid pharmacokinetics after oral single dose. J Pharmacokinet Pharmacodyn 2013; 40:479-86. [DOI: 10.1007/s10928-013-9323-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 06/07/2013] [Indexed: 01/23/2023]
|
41
|
Choi JR, Hong SW, Kim Y, Jang SE, Kim NJ, Han MJ, Kim DH. Metabolic activities of ginseng and its constituents, ginsenoside rb1 and rg1, by human intestinal microflora. J Ginseng Res 2013; 35:301-7. [PMID: 23717073 PMCID: PMC3659535 DOI: 10.5142/jgr.2011.35.3.301] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/17/2011] [Accepted: 05/17/2011] [Indexed: 11/29/2022] Open
Abstract
To evaluate the difference in expressing pharmacological effects of ginseng by intestinal microflora between Koreans, metabolic activities of ginseng, ginsenoside Rb1 and Rg1 by 100 fecal specimens were measured. The β-glucosidase activity for p-nitrophenyl- β-D-glucopyranoside was 0 to 0.42 μmol/min/mg and its average activity (mean±SD) was 0.10±0.07 μmol/min/mg. The metabolic activities of ginsenosides Rb1 and Rg1 were 0.01 to 0.42 and 0.01 to 0.38 pmol/min/mg, respectively. Their average activities were 0.25±0.08 and 0.15±0.09 pmol/min/mg, respectively. The compound K-forming activities from ginsenoside Rb1 and ginseng extract were 0 to 0.11 and 0 to 0.02 pmol/min/mg, respectively. Their average compound K-forming activities were 0.24±0.09 pmol/min/ mg and 2.14±3.66 fmol/min/mg, respectively. These activities all were not different between males and females, or between ages. Although compound K-forming activity from the aqueous extract of ginseng was low compared to that from ginenoside Rb1, their profiles were similar to those of isolated compounds. Based on these findings, we believe that the intestinal bacterial metabolic activities of ginseng components are variable in individuals and may be used as selection markers for responders to ginseng.
Collapse
Affiliation(s)
- Jong-Ryul Choi
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | | | |
Collapse
|
42
|
Comparative analysis of the gut microbiota in people with different levels of ginsenoside Rb1 degradation to compound K. PLoS One 2013; 8:e62409. [PMID: 23638073 PMCID: PMC3639287 DOI: 10.1371/journal.pone.0062409] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 03/21/2013] [Indexed: 12/25/2022] Open
Abstract
Panax ginseng (family Araliaceae) which contains ginsenoside Rb1 as a main constituent is traditionally used as a remedy for cancer, inflammation, stress, and ageing. The ginsenoside Rb1 in orally administered ginseng is metabolized to bioactive compounds by gut microbiota before their absorptions to the blood. However, its metabolizing activities in individuals are significantly different as we previously demonstrated. Here, we selected 5 samples with fecal activity potently metabolizing ginsenoside Rb1 to compound K (FPG; metabolic activity, 0.058±0.029 pmol/min/mg) and 5 samples with fecal activity non-metabolizing ginsenoside Rb1 to compound K (FNG) from a pool of 100 subjects investigated in a previous study and analyzed fecal microbiota by 16S rRNA gene pyrosequencing. Taxonomy-based analysis showed that the population levels of Firmicutes and Proteobacteria in FPG were lower than in FNG, but those of Bacteroidetes and Tenericutes in FPG were higher than in FNG. At the genus level, the population levels of Clostridiales_uc_g, Oscillibacter, Ruminococcus, Holdemania, and Sutterella in FPG were significantly higher than in FNG, but that of Leuconostoc in FPG was lower than in FNG. The population levels of Bacteroides and Bifidobacterium, which potently metabolizes ginsenoside Rb1 to compound K were dramatically increased in FPG. The gut microbiota compositions of FPG and FNG were segregated on PCO2 by Principal Coordinate Analysis. Intestinal bacterial metabolism of ginseng, particularly ginsenoside Rb1, may be dependent on the composition of gut microbiota, such as Ruminococcus spp., Bacteroides spp. and Bifidobacterium spp.
Collapse
|
43
|
Effects of probiotic yoghurt on symptoms and intestinal microbiota in patients with irritable bowel syndrome. INT J DAIRY TECHNOL 2013. [DOI: 10.1111/1471-0307.12028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
Omwancha WS, Mallipeddi R, Valle BL, Neau SH. Chitosan as a pore former in coated beads for colon specific drug delivery of 5-ASA. Int J Pharm 2012. [PMID: 23200955 DOI: 10.1016/j.ijpharm.2012.11.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A multiparticulate product for colon-specific delivery of a small molecule drug has been developed and characterized. Microcrystalline cellulose core beads containing 5-aminosalicylic acid produced by extrusion-spheronization were coated with chitosan and Aquacoat(®) ECD mixtures according to a factorial design. Coated beads were characterized in terms of drug release, shape, and friability. The optimum formulation was enteric coated and exposed to media simulating conditions in the stomach, small intestine, and colon. Release studies in simulated intestinal fluid revealed that the drug release rate from the coated beads, which were spherical and rugged, depended on the level of chitosan in the coat and the coat thickness. Enlarged pores observed on the surface of the coated beads exposed to the medium containing rat cecal and colonic enzymes are believed to have caused a significant enhancement of the drug release rate compared to the control exposed only to simulated gastric and intestinal fluids. The release mechanisms involved polymer relaxation and dissolved drug diffusion for simulated intestinal fluid and simulated colonic fluid, respectively. From the facilitated drug release in a colonic environment and the inhibition of drug release under gastric and intestinal conditions, it can be concluded that this multiparticulate system demonstrates the potential for colon-specific drug delivery.
Collapse
Affiliation(s)
- Wycliffe S Omwancha
- Philadelphia College of Pharmacy, University of the Sciences, 600 S. 43rd Street, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
45
|
Karthikkumar V, Sivagami G, Vinothkumar R, Rajkumar D, Nalini N. Modulatory efficacy of rosmarinic acid on premalignant lesions and antioxidant status in 1,2-dimethylhydrazine induced rat colon carcinogenesis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 34:949-58. [PMID: 22960260 DOI: 10.1016/j.etap.2012.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 07/20/2012] [Accepted: 07/23/2012] [Indexed: 06/01/2023]
Abstract
Colorectal cancer is one of the leading causes of cancer related deaths in Western countries and is becoming increasingly common in Asia. Rosmarinic acid (RA), one of the major components of polyphenol possesses attractive remedial features. The purpose of this study is to investigate the possible chemopreventive mechanism of action of RA against 1,2-dimethylhydrazine (DMH) induced rat colon carcinogenesis by evaluating the circulatory antioxidant status and colonic bacterial enzymes activities. Additionally, we analyzed the aberrant crypt foci (ACF) formation and multiplicity in the colon of experimental groups. Wistar male rats were divided into six groups. Group 1 was control rats, group 2 rats received RA (10 mg/kg b.w., p.o. everyday), rats in groups 3-6 received DMH (20 mg/kg b.w., s.c.) for the first 4 weeks. In addition to DMH, groups 4-6 received 2.5, 5, and 10 mg/kg b.w. RA respectively. The results revealed that supplementation with RA significantly reduced the formation of ACF and ACF multiplicity in DMH treated rats. Moreover RA supplementation prevented the alterations in circulatory antioxidant enzymes and colonic bacterial enzymes activities. Overall, our results showed that all three doses of RA inhibited carcinogenesis, though the effect of the intermediary dose of 5 mg/kg b.w. was more pronounced.
Collapse
Affiliation(s)
- V Karthikkumar
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar 608 002, Tamilnadu, India
| | | | | | | | | |
Collapse
|
46
|
Shrivastava R, Kannan A, Upreti RK, Chaturvedi UC. Effects of chromium on the resident gut bacteria of rat. Toxicol Mech Methods 2012; 15:211-8. [PMID: 20021085 DOI: 10.1080/15376520590945630] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The major nonoccupational source of chromium (Cr) for humans is through ingestion with food and water, but its effect on the gut microflora has not been studied. The present study was, therefore, undertaken to investigate the effects of chronic ingestion of potassium dichromate (chromium VI) on the resident gut bacteria of male Wistar rats. A group of rats was kept on drinking water containing 10 ppm chromium VI (Cr [VI]) (called Cr-stressed animals) and the other group was given plain water. After 10 weeks, Lactobacillus, Pseudomonas sp., and Escherichia coli were isolated from the cecum of the rats and various studies were performed. The most significant findings of the present study were the stimulation of growth of facultative gut bacteria from the Cr-stressed rats and the significant increase of growth even in the presence of lower concentrations of Cr. Furthermore, the capacity to reduce Cr (VI) was significantly decreased along with the increased tolerance of the bacteria to Cr (higher minimum inhibitory concentration [MIC] values), which was associated with the development of antibiotic resistance. The effects were most marked with the Pseudomonas sp. and least with the E. coli. The antibiotic resistance developed with the Lactobacillus may be a blessing in disguise, because the bacteria may continue to provide benefits even in patients given antibiotic therapy. The gut bacteria thus provide the first line of defense to the body by converting toxic Cr (VI) to a less toxic Cr (III) and may act as a prebiotic.
Collapse
Affiliation(s)
- Richa Shrivastava
- Biomembrane Division, Industrial Toxicology Research Centre, Mahatma Gandhi Marg, LucknowIndia
| | | | | | | |
Collapse
|
47
|
Jang SE, Jung IH, Joh EH, Han MJ, Kim DH. Antibiotics attenuate anti-scratching behavioral effect of ginsenoside Re in mice. JOURNAL OF ETHNOPHARMACOLOGY 2012; 142:105-112. [PMID: 22855946 DOI: 10.1016/j.jep.2012.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The root of Panax ginseng CA Meyer (ginseng) has been used for diabetes, cancer, stress and allergic diseases in the traditional Chinese medicine. AIM OF THE STUDY To understand the role of intestinal microflora in the pharmacological effect of ginsenoside Re, which is a main constituent of ginseng, we investigated its anti-scratching behavioral effect in the mice treated with or without antibiotics. MATERIALS AND METHODS Ginsenoside Re was orally administered to the mice treated with antibiotics (cefadroxil, oxytetracycline and erythromycin mixture (COE), streptomycin or/and tetracycline) and then investigated the relationship between ginsenoside Re-metabolizing β-glucosidase and α-rhamnosidase activities of intestinal microflora and its antiscratching behavioral effect. The anti-scratching behavioral effects of ginsenosides were investigated in the increments of 1 h and 6 h after their oral administrations. The scratching behavioral frequency was measured for 1 h after treatment with histamine. RESULTS Ginsenoside Re inhibited histamine-induced scratching behavior in mice. The anti-scratching behavioral effect of ginsenoside Re was more potent 6 h after its oral administration than 1 h after. However, its inhibitory effect was significantly attenuated in mice treated with COE, but it nearly was not affected in mice treated with streptomycin and/or tetracycline. Treatment with COE also significantly lowered fecal ginsenoside Re-metabolizing β-glucosidase and α-rhamnosidase activities in mice, as well as fecal metabolic activity of ginsenoside Re to ginsenoside Rh1. The anti-scratching behavioral effect of ginsenoside Rh1, a metabolite of ginsenoside Re by intestinal microflora, was superior to that of ginsenoside Re. Ginsenoside Rh1 potently inhibited the expression of IL-4 and TNF-α, as well as the activation of NF-κB and c-jun activation in histamine-stimulated scratching behavioral mice. CONCLUSION Ginsenoside Re may be metabolized to ginsenoside Rh1 by intestinal microflora, which enhances its anti-scratching behavioral effect by inhibiting NF-κB and c-jun activations.
Collapse
Affiliation(s)
- Se-Eun Jang
- Department of Food and Nutrition, Kyung Hee University, 1 Hoegi, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | | | | | | | | |
Collapse
|
48
|
Pan H, Feng J, He GX, Cerniglia CE, Chen H. Evaluation of impact of exposure of Sudan azo dyes and their metabolites on human intestinal bacteria. Anaerobe 2012; 18:445-53. [PMID: 22634331 DOI: 10.1016/j.anaerobe.2012.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 04/30/2012] [Accepted: 05/09/2012] [Indexed: 02/07/2023]
Abstract
Sudan azo dyes are banned for food usage in most countries, but they are illegally used to maintain or enhance the color of food products due to low cost, bright staining, and wide availability of the dyes. In this report, we examined the toxic effects of these azo dyes and their potential reduction metabolites on 11 prevalent human intestinal bacterial strains. Among the tested bacteria, cell growth of 2, 3, 5, 5, and 1 strains was inhibited by Sudan I, II, III, IV, and Para Red, respectively. At the tested concentration of 100 μM, Sudan I and II inhibited growth of Clostridium perfringens and Lactobacillus rhamnosus with decrease of growth rates from 14 to 47%. Sudan II also affected growth of Enterococcus faecalis. Growth of Bifidobacterium catenulatum, C. perfringens, E. faecalis, Escherichia coli, and Peptostreptococcus magnus was affected by Sudan III and IV with decrease in growth rates from 11 to 67%. C. perfringens was the only strain in which growth was affected by Para Red with 47 and 26% growth decreases at 6 and 10 h, respectively. 1-Amino-2-naphthol, a common metabolite of the dyes, was capable of inhibiting growth of most of the tested bacteria with inhibition rates from 8 to 46%. However, the other metabolites of the dyes had no effect on growth of the bacterial strains. The dyes and their metabolites had less effect on cell viability than on cell growth of the tested bacterial strains. Clostridium indolis and Clostridium ramosum were the only two strains with about a 10 % decrease in cell viability in the presence of Sudan azo dyes. The present results suggested that Sudan azo dyes and their metabolites potentially affect the human intestinal bacterial ecology by selectively inhibiting some bacterial species, which may have an adverse effect on human health.
Collapse
Affiliation(s)
- Hongmiao Pan
- Division of Microbiology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | | | | | | | | |
Collapse
|
49
|
Ritchie ML, Romanuk TN. A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLoS One 2012; 7:e34938. [PMID: 22529959 PMCID: PMC3329544 DOI: 10.1371/journal.pone.0034938] [Citation(s) in RCA: 315] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/11/2012] [Indexed: 12/12/2022] Open
Abstract
Background Meta-analyses on the effects of probiotics on specific gastrointestinal diseases have generally shown positive effects on disease prevention and treatment; however, the relative efficacy of probiotic use for treatment and prevention across different gastrointestinal diseases, with differing etiology and mechanisms of action, has not been addressed. Methods/Principal Findings We included randomized controlled trials in humans that used a specified probiotic in the treatment or prevention of Pouchitis, Infectious diarrhea, Irritable Bowel Syndrome, Helicobacter pylori, Clostridium difficile Disease, Antibiotic Associated Diarrhea, Traveler's Diarrhea, or Necrotizing Enterocolitis. Random effects models were used to evaluate efficacy as pooled relative risks across the eight diseases as well as across probiotic species, single vs. multiple species, patient ages, dosages, and length of treatment. Probiotics had a positive significant effect across all eight gastrointestinal diseases with a relative risk of 0.58 (95% (CI) 0.51–0.65). Six of the eight diseases: Pouchitis, Infectious diarrhea, Irritable Bowel Syndrome, Helicobacter pylori, Clostridium difficile Disease, and Antibiotic Associated Diarrhea, showed positive significant effects. Traveler's Diarrhea and Necrotizing Enterocolitis did not show significant effects of probiotcs. Of the 11 species and species mixtures, all showed positive significant effects except for Lactobacillus acidophilus, Lactobacillus plantarum, and Bifidobacterium infantis. Across all diseases and probiotic species, positive significant effects of probiotics were observed for all age groups, single vs. multiple species, and treatment lengths. Conclusions/Significance Probiotics are generally beneficial in treatment and prevention of gastrointestinal diseases. Efficacy was not observed for Traveler's Diarrhea or Necrotizing Enterocolitis or for the probiotic species L. acidophilus, L. plantarum, and B. infantis. When choosing to use probiotics in the treatment or prevention of gastrointestinal disease, the type of disease and probiotic species (strain) are the most important factors to take into consideration.
Collapse
Affiliation(s)
- Marina L Ritchie
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | |
Collapse
|
50
|
Smith AR, Macfarlane GT, Reynolds N, O'May GA, Bahrami B, Macfarlane S. Effect of a synbiotic on microbial community structure in a continuous culture model of the gastric microbiota in enteral nutrition patients. FEMS Microbiol Ecol 2012; 80:135-45. [DOI: 10.1111/j.1574-6941.2011.01279.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 12/02/2011] [Accepted: 12/05/2011] [Indexed: 01/22/2023] Open
Affiliation(s)
- Aileen R. Smith
- Microbiology and Gut Biology Group; University of Dundee; Ninewells Hospital and Medical School; Dundee; UK
| | - George T. Macfarlane
- Microbiology and Gut Biology Group; University of Dundee; Ninewells Hospital and Medical School; Dundee; UK
| | - Nigel Reynolds
- Department of Gastroenterology; Ninewells Hospital and Medical School; Dundee; UK
| | - Graeme A. O'May
- Microbiology and Gut Biology Group; University of Dundee; Ninewells Hospital and Medical School; Dundee; UK
| | - Bahram Bahrami
- Microbiology and Gut Biology Group; University of Dundee; Ninewells Hospital and Medical School; Dundee; UK
| | - Sandra Macfarlane
- Microbiology and Gut Biology Group; University of Dundee; Ninewells Hospital and Medical School; Dundee; UK
| |
Collapse
|