1
|
Deyaert S, Moens F, Pirovano W, van den Bogert B, Klaassens ES, Marzorati M, Van de Wiele T, Kleerebezem M, Van den Abbeele P. Development of a reproducible small intestinal microbiota model and its integration into the SHIME®-system, a dynamic in vitro gut model. Front Microbiol 2023; 13:1054061. [PMID: 37008301 PMCID: PMC10063983 DOI: 10.3389/fmicb.2022.1054061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/14/2022] [Indexed: 03/19/2023] Open
Abstract
The human gastrointestinal tract consists of different regions, each characterized by a distinct physiology, anatomy, and microbial community. While the colonic microbiota has received a lot of attention in recent research projects, little is known about the small intestinal microbiota and its interactions with ingested compounds, primarily due to the inaccessibility of this region in vivo. This study therefore aimed to develop and validate a dynamic, long-term simulation of the ileal microbiota using the SHIME®-technology. Essential parameters were identified and optimized from a screening experiment testing different inoculation strategies, nutritional media, and environmental parameters over an 18-day period. Subjecting a synthetic bacterial consortium to the selected conditions resulted in a stable microbiota that was representative in terms of abundance [8.81 ± 0.12 log (cells/ml)], composition and function. Indeed, the observed community mainly consisted of the genera Streptococcus, Veillonella, Enterococcus, Lactobacillus, and Clostridium (qPCR and 16S rRNA gene targeted Illumina sequencing), while nutrient administration boosted lactate production followed by cross-feeding interactions towards acetate and propionate. Furthermore, similarly as in vivo, bile salts were only partially deconjugated and only marginally converted into secondary bile salts. After confirming reproducibility of the small intestinal microbiota model, it was integrated into the established M-SHIME® where it further increased the compositional relevance of the colonic community. This long-term in vitro model provides a representative simulation of the ileal bacterial community, facilitating research of the ileum microbiota dynamics and activity when, for example, supplemented with microbial or diet components. Furthermore, integration of this present in vitro simulation increases the biological relevance of the current M-SHIME® technology.
Collapse
Affiliation(s)
| | | | | | | | | | - Massimo Marzorati
- ProDigest BV, Gent, Belgium
- Center of Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
- *Correspondence: Massimo Marzorati,
| | - Tom Van de Wiele
- ProDigest BV, Gent, Belgium
- Center of Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Gent, Belgium
| | - Michiel Kleerebezem
- Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | | |
Collapse
|
2
|
Song I, Gotoh Y, Ogura Y, Hayashi T, Fukiya S, Yokota A. Comparative Genomic and Physiological Analysis against Clostridium scindens Reveals Eubacterium sp. c-25 as an Atypical Deoxycholic Acid Producer of the Human Gut Microbiota. Microorganisms 2021; 9:microorganisms9112254. [PMID: 34835380 PMCID: PMC8623032 DOI: 10.3390/microorganisms9112254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/27/2022] Open
Abstract
The human gut houses bile acid 7α-dehydroxylating bacteria that produce secondary bile acids such as deoxycholic acid (DCA) from host-derived bile acids through enzymes encoded by the bai operon. While recent metagenomic studies suggest that these bacteria are highly diverse and abundant, very few DCA producers have been identified. Here, we investigated the physiology and determined the complete genome sequence of Eubacterium sp. c-25, a DCA producer that was isolated from human feces in the 1980s. Culture experiments showed a preference for neutral to slightly alkaline pH in both growth and DCA production. Genomic analyses revealed that c-25 is phylogenetically distinct from known DCA producers and possesses a multi-cluster arrangement of predicted bile-acid inducible (bai) genes that is considerably different from the typical bai operon structure. This arrangement is also found in other intestinal bacterial species, possibly indicative of unconfirmed 7α-dehydroxylation capabilities. Functionality of the predicted bai genes was supported by the induced expression of baiB, baiCD, and baiH in the presence of cholic acid substrate. Taken together, Eubacterium sp. c-25 is an atypical DCA producer with a novel bai gene cluster structure that may represent an unexplored genotype of DCA producers in the human gut.
Collapse
Affiliation(s)
- Isaiah Song
- Laboratory of Microbial Physiology, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan; (I.S.); (A.Y.)
| | - Yasuhiro Gotoh
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.G.); (T.H.)
| | - Yoshitoshi Ogura
- Department of Infectious Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan;
| | - Tetsuya Hayashi
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (Y.G.); (T.H.)
| | - Satoru Fukiya
- Laboratory of Microbial Physiology, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan; (I.S.); (A.Y.)
- Correspondence: ; Tel.: +81-11-706-2501
| | - Atsushi Yokota
- Laboratory of Microbial Physiology, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan; (I.S.); (A.Y.)
| |
Collapse
|
3
|
Bozward AG, Ronca V, Osei-Bordom D, Oo YH. Gut-Liver Immune Traffic: Deciphering Immune-Pathogenesis to Underpin Translational Therapy. Front Immunol 2021; 12:711217. [PMID: 34512631 PMCID: PMC8425300 DOI: 10.3389/fimmu.2021.711217] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
The tight relationship between the gut and liver on embryological, anatomical and physiological levels inspired the concept of a gut-liver axis as a central element in the pathogenesis of gut-liver axis diseases. This axis refers to the reciprocal regulation between these two organs causing an integrated system of immune homeostasis or tolerance breakdown guided by the microbiota, the diet, genetic background, and environmental factors. Continuous exposure of gut microbiome, various hormones, drugs and toxins, or metabolites from the diet through the portal vein adapt the liver to maintain its tolerogenic state. This is orchestrated by the combined effort of immune cells network: behaving as a sinusoidal and biliary firewall, along with a regulatory network of immune cells including, regulatory T cells and tolerogenic dendritic cells (DC). In addition, downregulation of costimulatory molecules on hepatic sinusoids, hepatocytes and biliary epithelial cells as well as regulating the bile acids chain also play a part in hepatic immune homeostasis. Recent evidence also demonstrated the link between changes in the gut microbiome and liver resident immune cells in the progression of cirrhosis and the tight correlation among primary sclerosing cholangitis (PSC) and also checkpoint induced liver and gut injury. In this review, we will summarize the most recent evidence of the bidirectional relationship among the gut and the liver and how it contributes to liver disease, focusing mainly on PSC and checkpoint induced hepatitis and colitis. We will also focus on completed therapeutic options and on potential targets for future treatment linking with immunology and describe the future direction of this research, taking advantage of modern technologies.
Collapse
Affiliation(s)
- Amber G. Bozward
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Transplant and Hepatobiliary Unit, Queen Elizabeth Hospital, University Hospital of Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network - Rare Liver Centre, Birmingham, United Kingdom
- Birmingham Advanced Cellular Therapy Facility, University of Birmingham, Birmingham, United Kingdom
| | - Vincenzo Ronca
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Transplant and Hepatobiliary Unit, Queen Elizabeth Hospital, University Hospital of Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network - Rare Liver Centre, Birmingham, United Kingdom
| | - Daniel Osei-Bordom
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Queen Elizabeth Hospital, University Hospital of Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
| | - Ye Htun Oo
- Centre for Liver and Gastrointestinal Research and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Transplant and Hepatobiliary Unit, Queen Elizabeth Hospital, University Hospital of Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network - Rare Liver Centre, Birmingham, United Kingdom
- Birmingham Advanced Cellular Therapy Facility, University of Birmingham, Birmingham, United Kingdom
- Queen Elizabeth Hospital, University Hospital of Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
4
|
The Role of Microbiota in Primary Sclerosing Cholangitis and Related Biliary Malignancies. Int J Mol Sci 2021; 22:ijms22136975. [PMID: 34203536 PMCID: PMC8268159 DOI: 10.3390/ijms22136975] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 02/08/2023] Open
Abstract
Primary sclerosing cholangitis (PSC) is an immune-related cholangiopathy characterized by biliary inflammation, cholestasis, and multifocal bile duct strictures. It is associated with high rates of progression to end-stage liver disease as well as a significant risk of cholangiocarcinoma (CCA), gallbladder cancer, and colorectal carcinoma. Currently, no effective medical treatment with an impact on the overall survival is available, and liver transplantation is the only curative treatment option. Emerging evidence indicates that gut microbiota is associated with disease pathogenesis. Several studies analyzing fecal and mucosal samples demonstrate a distinct gut microbiome in individuals with PSC compared to healthy controls and individuals with inflammatory bowel disease (IBD) without PSC. Experimental mouse and observational human data suggest that a diverse set of microbial functions may be relevant, including microbial metabolites and bacterial processing of pharmacological agents, bile acids, or dietary compounds, altogether driving the intrahepatic inflammation. Despite critical progress in this field over the past years, further functional characterization of the role of the microbiota in PSC and related malignancies is needed. In this review, we discuss the available data on the role of the gut microbiome and elucidate important insights into underlying pathogenic mechanisms and possible microbe-altering interventions.
Collapse
|
5
|
Tsujimoto H, Hirata Y, Ueda Y, Kinoshita N, Tawa H, Tanaka Y, Koshiba R, Ota K, Kojima Y, Kakimoto K, Takeuchi T, Miyazaki T, Nakamura S, Higuchi K. Effect of a proton-pump inhibitor on intestinal microbiota in patients taking low-dose aspirin. Eur J Clin Pharmacol 2021; 77:1639-1648. [PMID: 34085115 DOI: 10.1007/s00228-021-03167-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIM Low-dose aspirin (LDA) administration prevents cerebral infarction and myocardial infarction, but many studies found an association with mucosal injury. Proton-pump inhibitors (PPIs) can prevent gastric and duodenal mucosal damage, but they may exacerbate small-intestinal mucosal injury by altering the microbiota. We aimed to assess the effect of PPIs on the intestinal flora of LDA users. METHODS Thirty-two recruited patients, who received LDA (100 mg/day) but did not take PPIs, were divided into 15 patients additionally receiving esomeprazole (20 mg/day) and 17 patients additionally receiving vonoprazan (10 mg/day). On days 0, 30, 90, and 180, the microbiota of each patient was examined by terminal restriction fragment length polymorphism analysis, and the serum gastrin, hemoglobin, and hematocrit levels were measured. RESULTS Additional PPI administration increased the proportion of Lactobacillales in the microbiota of LDA users. This trend was more prevalent in the vonoprazan group (p < 0.0001) than in the esomeprazole group (p = 0.0024). The Lactobacillales proportion was positively correlated with the gastrin level (r = 0.5354). No significant hemoglobin or hematocrit level reduction was observed in subjects receiving LDA with additional PPI. CONCLUSIONS Additional PPI administration increased the Lactobacillales proportion in the microbiota of LDA users. The positive correlation between the gastrin level and the proportion of Lactobacillales suggested that the change in the intestinal flora was associated with the degree of suppression of gastric acid secretion. Additional oral PPI did not significantly promote anemia, but the risk of causing PPI-induced small-intestinal mucosal injury in LDA users should be considered.
Collapse
Affiliation(s)
- Hiroyuki Tsujimoto
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Yuki Hirata
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan.
| | - Yasuhiro Ueda
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Naohiko Kinoshita
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Hideki Tawa
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Yasuyoshi Tanaka
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Ryoji Koshiba
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Kazuhiro Ota
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Yuichi Kojima
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Kazuki Kakimoto
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Toshihisa Takeuchi
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Takako Miyazaki
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Shiro Nakamura
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| | - Kazuhide Higuchi
- Second Department of Internal Medicine, Osaka Medical College, 2-7 Daigakumachi, Takatsuki City, Osaka, 569-8686, Japan
| |
Collapse
|
6
|
Abstract
Primary sclerosing cholangitis (PSC) is a rare, immune-mediated, chronic cholestatic liver disease associated with a unique phenotype of inflammatory bowel disease that frequently manifests as pancolitis with right-sided predominance. Available data suggest a bidirectional interplay of the gut-liver axis with critical roles for the gastrointestinal microbiome and circulating bile acids (BAs) in the pathophysiology of PSC. BAs shape the gut microbiome, whereas gut microbes have the potential to alter BAs, and there are emerging data that alterations of BAs and the microbiome are not simply a consequence but the cause of PSC. Clustering of PSC in families may suggest that PSC occurs in genetically susceptible individuals. After exposure to an environmental trigger (e.g., microbial byproducts or BAs), an aberrant or exaggerated cholangiocyte-induced immune cascade occurs, ultimately leading to bile duct damage and progressive fibrosis. The pathophysiology can be conceptualized as a triad of (1) gut dysbiosis, (2) altered BA metabolism, and (3) immune-mediated biliary injury. Immune activation seems to be central to the disease process, but immunosuppression does not improve clinical outcomes or alter the natural history of PSC. Currently, orthoptic liver transplantation is the only established life-saving treatment, whereas antimicrobial therapy or fecal transplantation is an emerging therapeutic option for PSC. The beneficial effects of these microbiome-based therapies are likely mediated by a shift of the gut microbiome with favorable effects on BA metabolism. In the future, personalized approaches will allow to better target the interdependence between microbiome, immune function, and BA metabolism and potentially cure patients with PSC.
Collapse
|
7
|
Somm E, Henry H, Bruce SJ, Bonnet N, Montandon SA, Niederländer NJ, Messina A, Aeby S, Rosikiewicz M, Fajas L, Sempoux C, Ferrari SL, Greub G, Pitteloud N. β-Klotho deficiency shifts the gut-liver bile acid axis and induces hepatic alterations in mice. Am J Physiol Endocrinol Metab 2018; 315:E833-E847. [PMID: 29944388 DOI: 10.1152/ajpendo.00182.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
β-Klotho (encoded by Klb) is an obligate coreceptor, mediating both fibroblast growth factor (FGF)15 and FGF21 signaling. Klb-/- mice are refractory to metabolic FGF15 and FGF21 action and exhibit derepressed (increased) bile acid (BA) synthesis. Here, we deeply phenotyped male Klb-/- mice on a pure C57BL/6J genetic background, fed a chow diet focusing on metabolic aspects. This aims to better understand the physiological consequences of concomitant FGF15 and FGF21 signaling deficiency, in particular on the gut-liver axis. Klb-/- mice present permanent growth restriction independent of adiposity and energy balance. Klb-/- mice also exhibit few changes in carbohydrate metabolism, combining normal gluco-tolerance, insulin sensitivity, and fasting response with increased gluconeogenic capacity and decreased glycogen mobilization. Livers of Klb-/- mice reveal pathologic features, including a proinflammatory status and initiation of fibrosis. These defects are associated to a massive shift in BA composition in the enterohepatic system and blood circulation featured by a large excess of microbiota-derived deoxycholic acid, classically known for its genotoxicity in the gastrointestinal tract. In conclusion, β-Klotho is a gatekeeper of hepatic integrity through direct action (mediating FGF21 anti-inflammatory signaling) and indirect mechanisms (mediating FGF15 signaling that maintains BA level and composition).
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Hugues Henry
- Clinical Chemistry Laboratory, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Stephen J Bruce
- Clinical Chemistry Laboratory, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Nicolas Bonnet
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Sophie A Montandon
- Service of Endocrinology, Diabetes, Hypertension, and Nutrition, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Nicolas J Niederländer
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Andrea Messina
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Sébastien Aeby
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Marta Rosikiewicz
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Lluis Fajas
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne , Lausanne , Switzerland
| | - Christine Sempoux
- Institute of Pathology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Serge L Ferrari
- Division of Bone Diseases, Department of Internal Medicine Specialties, Geneva University Hospital and Faculty of Medicine , Geneva , Switzerland
| | - Gilbert Greub
- Institute of Microbiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology, and Metabolism, Department of Physiology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne , Lausanne , Switzerland
| |
Collapse
|
8
|
Del Chierico F, Abbatini F, Russo A, Quagliariello A, Reddel S, Capoccia D, Caccamo R, Ginanni Corradini S, Nobili V, De Peppo F, Dallapiccola B, Leonetti F, Silecchia G, Putignani L. Gut Microbiota Markers in Obese Adolescent and Adult Patients: Age-Dependent Differential Patterns. Front Microbiol 2018; 9:1210. [PMID: 29922272 PMCID: PMC5996250 DOI: 10.3389/fmicb.2018.01210] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/17/2018] [Indexed: 12/24/2022] Open
Abstract
Obesity levels, especially in children, have dramatically increased over the last few decades. Recently, several studies highlighted the involvement of gut microbiota in the pathophysiology of obesity. We investigated the composition of gut microbiota in obese adolescents and adults compared to age-matched normal weight (NW) volunteers in order to assemble age- and obesity-related microbiota profiles. The composition of gut microbiota was analyzed by 16S rRNA-based metagenomics. Ecological representations of microbial communities were computed, and univariate, multivariate, and correlation analyses performed on bacterial profiles. The prediction of metagenome functional content from 16S rRNA gene surveys was carried out. Ecological analyses revealed a dissimilarity among the subgroups, and resultant microbiota profiles differed between obese adolescents and adults. Using statistical analyses, we assigned, as microbial markers, Faecalibacterium prausnitzii and Actinomyces to the microbiota of obese adolescents, and Parabacteroides, Rikenellaceae, Bacteroides caccae, Barnesiellaceae, and Oscillospira to the microbiota of NW adolescents. The predicted metabolic profiles resulted different in adolescent groups. Particularly, biosynthesis of primary bile acid and steroid acids, metabolism of fructose, mannose, galactose, butanoate, and pentose phosphate and glycolysis/gluconeogenesis were for the majority associated to obese, while biosynthesis and metabolism of glycan, biosynthesis of secondary bile acid, metabolism of steroid hormone and lipoic acid were associated to NW adolescents. Our study revealed unique features of gut microbiota in terms of ecological patterns, microbial composition and metabolism in obese patients. The assignment of novel obesity bacterial markers may open avenues for the development of patient-tailored treatments dependent on age-related microbiota profiles.
Collapse
Affiliation(s)
| | - Francesca Abbatini
- Department of Medical Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, Bariatric Center of Excellence IFSO-EU, Sapienza University, Rome, Italy
| | - Alessandra Russo
- Human Microbiome Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Sofia Reddel
- Human Microbiome Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Danila Capoccia
- Department of Experimental Medicine, Section of Endocrinology, Sapienza University, Rome, Italy
| | - Romina Caccamo
- Pediatric General Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Valerio Nobili
- Department of Pediatrics, Sapienza University, Rome, Italy.,Hepatogastroenterology and Nutrition Unit, Bambino Gesù Hospital, Rome, Italy
| | - Francesco De Peppo
- Pediatric General Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Bruno Dallapiccola
- Scientific Directorate, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Frida Leonetti
- Department of Experimental Medicine, Section of Endocrinology, Sapienza University, Rome, Italy
| | - Gianfranco Silecchia
- Department of Medical Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, Bariatric Center of Excellence IFSO-EU, Sapienza University, Rome, Italy
| | - Lorenza Putignani
- Human Microbiome Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Parasitology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
9
|
Kawano A, Ishikawa H, Mutoh M, Kubota H, Matsuda K, Tsuji H, Matsumoto K, Nomoto K, Tanaka R, Nakamura T, Wakabayashi K, Sakai T. Higher enterococcus counts indicate a lower risk of colorectal adenomas: a prospective cohort study. Oncotarget 2018; 9:21459-21467. [PMID: 29765552 PMCID: PMC5940372 DOI: 10.18632/oncotarget.25130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/24/2018] [Indexed: 01/08/2023] Open
Abstract
Intestinal bacteria play an important role in human health. This prospective cohort study aimed to investigate the relationship between the abundance of different intestinal bacteria and the risk of developing colorectal cancer (CRC). Fecal samples from CRC patients (n = 157) were collected at the start of the study wherein patients subsequently underwent endoscopy to remove polyps. Gut bacteria were isolated by using specific culture methods and the fecal counts of various bacteria were quantified by reverse-transcription-quantitative-PCR (RT-qPCR) assays. The obtained data were subjected to cohort analysis in relation to the incidence of colorectal adenomas after 4 years of intervention. No relationship was detected between the counts of major intestinal bacteria and the incidence of colorectal adenomas. However, interestingly, a significant negative correlation was noted between colorectal adenoma incidence and the counts of bacteria grown on Columbia blood agar base (COBA) (P = 0.007). The risk ratio of colorectal adenomas was 0.58 (95% CI: 0.35–0.96) in the group with the highest bacterial count compared to the lowest. Bacteria grown on COBA were more abundant in older patients, non-smoking patients, and patients with a lower body mass index. The RT-qPCR results revealed a significantly lower colorectal adenoma incidence in subjects with higher enterococcal count as compared to subjects with a lower count, with a risk ratio of 0.47 (95% CI: 0.30–0.76). Correlation of a higher enterococci count with a lower risk of CRC development suggests that certain Enterococcus strains may have adenoma suppressive effects.
Collapse
Affiliation(s)
- Atsuko Kawano
- Institute of Gastroenterology, Zenjinkai Shimin-no-Mori Hospital, Miyazaki, Japan
| | - Hideki Ishikawa
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michihiro Mutoh
- Epidemiology and Prevention Group, Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo, Japan
| | | | - Kazunori Matsuda
- Yakult Honsha European Research Center for Microbiology, ESV, Gent Zwijnaarde, Belgium
| | | | | | | | | | | | - Keiji Wakabayashi
- Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Toshiyuki Sakai
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
10
|
Isolation of six novel 7-oxo- or urso-type secondary bile acid-producing bacteria from rat cecal contents. J Biosci Bioeng 2017; 124:514-522. [PMID: 28751127 DOI: 10.1016/j.jbiosc.2017.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/17/2017] [Accepted: 06/05/2017] [Indexed: 01/01/2023]
Abstract
Understanding the dynamics of secondary bile acid (SBA) formation in the gut by SBA-producing bacteria is important for host health, as SBAs have been shown to affect host pathophysiology and gut microbiota composition. However, our knowledge of SBA producers is limited in light of the diversity of gut microbes. Here, we isolated six novel SBA-producing bacteria from rat cecal contents, all of which were members of known species of gut microbes. Anaerostipes caccae D10, Bacteroides nordii C5, Clostridioides difficile D7, and Clostridium cadaveris G11 were capable of oxidizing cholic acid and chenodeoxycholic acid into 7-oxo-derivatives with varying yields. B. nordii C5 and its type strain JCM 12987T had the highest molar yield, ∼90%. Clostridium disporicum F4 and Clostridium subterminale C4 epimerized cholic acid into ursocholic acid with yields of ∼85%; the corresponding type strains lacked epimerization activity. Furthermore, although not novel as an SBA producer, Clostridium scindens G10 that produced deoxycholic acid from cholic acid was isolated for the first time from rodents. These findings will contribute to elucidation of SBA formation in the gut.
Collapse
|
11
|
Mobini R, Tremaroli V, Ståhlman M, Karlsson F, Levin M, Ljungberg M, Sohlin M, Bertéus Forslund H, Perkins R, Bäckhed F, Jansson PA. Metabolic effects of Lactobacillus reuteri DSM 17938 in people with type 2 diabetes: A randomized controlled trial. Diabetes Obes Metab 2017; 19:579-589. [PMID: 28009106 DOI: 10.1111/dom.12861] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/08/2016] [Accepted: 12/20/2016] [Indexed: 12/16/2022]
Abstract
AIMS To investigate the metabolic effects of 12-week oral supplementation with Lactobacillus reuteri DSM 17938 in patients with type 2 diabetes on insulin therapy. MATERIALS AND METHODS In a double-blind trial, we randomized 46 people with type 2 diabetes to placebo or a low (108 CFU/d) or high dose (1010 CFU/d) of L. reuteri DSM 17938 for 12 weeks. The primary endpoint was the effect of supplementation on glycated haemoglobin (HbA1c). Secondary endpoints were insulin sensitivity (assessed by glucose clamp), liver fat content, body composition, body fat distribution, faecal microbiota composition and serum bile acids. RESULTS Supplementation with L. reuteri DSM 17938 for 12 weeks did not affect HbA1c, liver steatosis, adiposity or microbiota composition. Participants who received the highest dose of L. reuteri exhibited increases in insulin sensitivity index (ISI) and serum levels of the secondary bile acid deoxycholic acid (DCA) compared with baseline, but these differences were not significant in the between-group analyses. Post hoc analysis showed that participants who responded with increased ISI after L. reuteri supplementation had higher microbial diversity at baseline, and increased serum levels of DCA after supplementation. In addition, increases in DCA levels correlated with improvement in insulin sensitivity in the probiotic recipients. CONCLUSIONS Intake of L. reuteri DSM 17938 for 12 weeks did not affect HbA1c in people with type 2 diabetes on insulin therapy; however, L. reuteri improved insulin sensitivity in a subset of participants and we propose that high diversity of the gut microbiota at baseline may be important.
Collapse
Affiliation(s)
- Reza Mobini
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Karlsson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Max Levin
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Maria Ljungberg
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maja Sohlin
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Heléne Bertéus Forslund
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rosie Perkins
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Centre for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Per-Anders Jansson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
12
|
Gu S, Cao B, Sun R, Tang Y, Paletta JL, Wu XL, Liu L, Zha W, Zhao C, Li Y, Radlon JM, Hylemon PB, Zhou H, Aa J, Wang G. A metabolomic and pharmacokinetic study on the mechanism underlying the lipid-lowering effect of orally administered berberine. MOLECULAR BIOSYSTEMS 2015; 11:463-74. [PMID: 25411028 PMCID: PMC4302037 DOI: 10.1039/c4mb00500g] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clinical and animal studies demonstrated that orally administered berberine had a distinct lipid-lowering effect. However, pharmacokinetic studies showed that berberine was poorly absorbed into the body so the levels of berberine in the blood and target tissues were far below the effective concentrations revealed. To probe the underlying mechanism, the effect of berberine on the biological system was studied on a high-fat-diet-induced hamster hyperlipidemia model. Our results showed that intragastrically-administered berberine was poorly absorbed into circulation and most berberine accumulated in gut content. Although the bioavailability of intragastrically administered berberine was much lower than that of intraperitoneally administered berberine, it had a stronger lipid-lowing effect, indicating that the gastrointestinal tract is a potential target for the hypolipidemic effect of berberine. A metabolomic study on both serum and gut content showed that orally administered berberine significantly regulated molecules involved in lipid metabolism, and increased the generation of bile acids in the hyperlipidemic model. DNA analysis revealed that the orally administered berberine modulated the gut microbiota, and berberine showed a significant inhibition of the 7α-dehydroxylation conversion of cholic acid to deoxycholic acid, indicating a decreased elimination of bile acids in the gut. However, in model hamsters, elevated bile acids failed to downregulate the expression and function of CYP7A1 in a negative feedback loop. It was suggested that the hypocholesterolemic effect of orally administered berberine involves modulating the turnover of bile acids and the farnesoid X receptor signal pathway.
Collapse
Affiliation(s)
- Shenghua Gu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
- College of Engineering, Peking University, Beijing 100871, China
- Center for Drug Evaluation and Research, Shanghai University of Traditional Chinese Medicine
| | - Bei Cao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Runbin Sun
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Yueqing Tang
- College of Engineering, Peking University, Beijing 100871, China
| | - Janice L. Paletta
- Center for Drug Evaluation and Research, Shanghai University of Traditional Chinese Medicine
| | - Xiao-Lei Wu
- College of Engineering, Peking University, Beijing 100871, China
| | - Linsheng Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Weibin Zha
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Chunyan Zhao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Li
- College of Engineering, Peking University, Beijing 100871, China
| | - Jason M. Radlon
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298, USA
| | - Phillip B. Hylemon
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298, USA
| | - Huiping Zhou
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, Virginia, 23298, USA
| | - Jiye Aa
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, Jiangsu Key laboratory of drug design and optimization, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
13
|
Vrieze A, Out C, Fuentes S, Jonker L, Reuling I, Kootte RS, van Nood E, Holleman F, Knaapen M, Romijn JA, Soeters MR, Blaak EE, Dallinga-Thie GM, Reijnders D, Ackermans MT, Serlie MJ, Knop FK, Holst JJ, van der Ley C, Kema IP, Zoetendal EG, de Vos WM, Hoekstra JBL, Stroes ES, Groen AK, Nieuwdorp M. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J Hepatol 2014; 60:824-31. [PMID: 24316517 DOI: 10.1016/j.jhep.2013.11.034] [Citation(s) in RCA: 399] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Obesity has been associated with changes in the composition and function of the intestinal microbiota. Modulation of the microbiota by antibiotics also alters bile acid and glucose metabolism in mice. Hence, we hypothesized that short term administration of oral antibiotics in humans would affect fecal microbiota composition and subsequently bile acid and glucose metabolism. METHODS In this single blinded randomized controlled trial, 20 male obese subjects with metabolic syndrome were randomized to 7 days of amoxicillin 500 mg t.i.d. or 7 days of vancomycin 500 mg t.i.d. At baseline and after 1 week of therapy, fecal microbiota composition (Human Intestinal Tract Chip phylogenetic microarray), fecal and plasma bile acid concentrations as well as insulin sensitivity (hyperinsulinemic euglycemic clamp using [6,6-(2)H2]-glucose tracer) were measured. RESULTS Vancomycin reduced fecal microbial diversity with a decrease of gram-positive bacteria (mainly Firmicutes) and a compensatory increase in gram-negative bacteria (mainly Proteobacteria). Concomitantly, vancomycin decreased fecal secondary bile acids with a simultaneous postprandial increase in primary bile acids in plasma (p<0.05). Moreover, changes in fecal bile acid concentrations were predominantly associated with altered Firmicutes. Finally, administration of vancomycin decreased peripheral insulin sensitivity (p<0.05). Amoxicillin did not affect any of these parameters. CONCLUSIONS Oral administration of vancomycin significantly impacts host physiology by decreasing intestinal microbiota diversity, bile acid dehydroxylation and peripheral insulin sensitivity in subjects with metabolic syndrome. These data show that intestinal microbiota, particularly of the Firmicutes phylum contributes to bile acid and glucose metabolism in humans. This trial is registered at the Dutch Trial Register (NTR2566).
Collapse
Affiliation(s)
- Anne Vrieze
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Carolien Out
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Susana Fuentes
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Lisanne Jonker
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Isaie Reuling
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Ruud S Kootte
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Els van Nood
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Frits Holleman
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Max Knaapen
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Johannes A Romijn
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Maarten R Soeters
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Ellen E Blaak
- Department of Human Metabolism, NUTRIM, School for Nutrition, Toxicology and Metabolism, Maastricht University, The Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Dorien Reijnders
- Department of Human Metabolism, NUTRIM, School for Nutrition, Toxicology and Metabolism, Maastricht University, The Netherlands
| | - Mariëtte T Ackermans
- Department of Clinical Chemistry, Laboratory of Endocrinology, Academic Medical Center, Amsterdam, The Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
| | - Filip K Knop
- Department of Internal Medicine, Gentofte Hospital, Hellerup, Denmark; NNF Center for Basic Metabolic Research, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Denmark
| | - Jenst J Holst
- NNF Center for Basic Metabolic Research, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, Denmark
| | - Claude van der Ley
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands; Department of Basic Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Joost B L Hoekstra
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Erik S Stroes
- Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Albert K Groen
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Max Nieuwdorp
- Department of Medicine, Academic Medical Center, Amsterdam, The Netherlands; Department of Vascular Medicine and Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Kawano A, Ishikawa H, Nakamura T, Kono K. Evaluation of epidemiological studies of intestinal bacteria that affected occurrence of colorectal cancer: studies of prevention of colorectal tumors by dairy products and lactic acid bacteria. Nihon Eiseigaku Zasshi 2010; 65:422-446. [PMID: 20508386 DOI: 10.1265/jjh.65.422] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Enviromental factors have been consistently associated with colon cancer risk. In particular, consumption of Western-style diet including red meat is the most widely accepted etiologic risk factor. It has been reported that dietary factors change the proportion of intestinal flora, and it also affects the composition of fecal bile acids and the intestinal activity of some mutagens. In addition, it was suggested that modulating the composition of intestinal flora may reduce the occurrence of colorectal cancer. In this review, we present the clinical studies on the association between intestinal flora and the risk of colorectal cancer that have been carried out to date. The clinical studies of intestinal bacteria related to colorectal cancer risk have not shown consistent results so far, compared with the accomplishments of some basic studies. On the other hand, it was suggested in some clinical studies that lactic acid bacteria reduce the occurrence of colorectal cancer.
Collapse
Affiliation(s)
- Atsuko Kawano
- Division of Preventive and Social Medicine, Department of Hygiene and Public Health, Osaka Medical College
| | | | | | | |
Collapse
|
15
|
Kibe R, Sakamoto M, Yokota H, Benno Y. Characterization of the inhabitancy of mouse intestinal bacteria (MIB) in rodents and humans by real-time PCR with group-specific primers. Microbiol Immunol 2008; 51:349-57. [PMID: 17446674 DOI: 10.1111/j.1348-0421.2007.tb03916.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mouse intestinal bacteria (MIB) is a new operational taxonomic unit (OTU) belonging to the Bacteroides subgroup in the Cytophaga-Flavobacter-Bacteroides (CFB) phylum recently found in the intestine of mice, rats and humans. However, their characters are still unknown since they have not yet been isolated by culture. To understand their habitat characteristics in intestinal tracts, the quantification assays of MIB were established using MIB group-specific primers. The MIB population in the intestine was evaluated as a percentage of the number of 16S rRNA gene copy of MIB. A real-time PCR assay using group specific primers showed the fluctuation of MIB inhabitancy and revealed that the MIB population in the small intestine of mice was significantly lower than the large intestinal contents. Moreover, MIB was found in human feces though the number was lower than in murine. This assay using group-specific primers revealed new information about host-preference of MIB.
Collapse
Affiliation(s)
- Ryoko Kibe
- Department of Veterinary Biochemistry, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | | | | | | |
Collapse
|
16
|
Booijink CCGM, Zoetendal EG, Kleerebezem M, de Vos WM. Microbial communities in the human small intestine: coupling diversity to metagenomics. Future Microbiol 2007; 2:285-95. [PMID: 17661703 DOI: 10.2217/17460913.2.3.285] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract is the main site where the conversion and absorption of food components takes place. The host-derived physiological processes and the residing microorganisms, especially in the small intestine, contribute to this nutrient supply. To circumvent sampling problems of the small intestine, several model systems have been developed to study microbial diversity and functionality in the small intestine. In addition, metagenomics offers novel possibilities to gain insight into the genetic potential and functional properties of these microbial communities. Here, an overview is presented of the most recent insights into the diversity and functionality of the microorganisms in the human gastrointestinal tract, with a focus on the small intestine.
Collapse
Affiliation(s)
- Carien C G M Booijink
- Wageningen Centre for Food Sciences, and Laboratory of Microbiology, Hesselink van Suchtelenweg 4, Wageningen, The Netherlands.
| | | | | | | |
Collapse
|
17
|
Elsayed S, Zhang K. Isolation and 16S ribosomal RNA gene sequence-based identification of Clostridium scindens from an intra-abdominal abscess. Anaerobe 2006; 12:13-6. [PMID: 16701607 DOI: 10.1016/j.anaerobe.2005.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2005] [Revised: 06/01/2005] [Accepted: 06/06/2005] [Indexed: 10/25/2022]
Abstract
Clostridium scindens has not been previously associated with human infection. We describe a case of an adolescent female with Crohn's disease presenting with a post-surgical intra-abdominal abscess from which this organism was isolated in pure culture. This is the first documented report of human infection caused by this micro-organism.
Collapse
Affiliation(s)
- Sameer Elsayed
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada.
| | | |
Collapse
|
18
|
Sakata S, Tonooka T, Ishizeki S, Takada M, Sakamoto M, Fukuyama M, Benno Y. Culture-independent analysis of fecal microbiota in infants, with special reference toBifidobacteriumspecies. FEMS Microbiol Lett 2005; 243:417-23. [PMID: 15686844 DOI: 10.1016/j.femsle.2005.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Revised: 12/24/2004] [Accepted: 01/03/2005] [Indexed: 11/18/2022] Open
Abstract
Fecal microbiota of 31 breast-fed, 26 mix-fed, and 11 bottle-fed infants were analyzed by using terminal restriction fragment length polymorphism (T-RFLP), and culture method. We first determined the total and cultivated bacterial counts in infant fecal microbiota. Only approximately 30% of bacteria present in fecal microbiota were cultivable while the remainder was yet-to-be cultured bacteria. Sixty-eight fecal samples were divided into two clusters (I and II) by T-RFLP analysis, and then subdivided into five subclusters (Ia, Ib, IIa, IIb and IIc). There was no clear relationship between clusters and feeding method. A proportion of bifidobacteria was detected in the fecal material by PCR method using species-specific primers. The predominant Bifidobacterium spp. was Bifidobacterium longum longum type (43 samples (63.2%)), followed by B. longum infantis type (23 samples (33.8%)) and B. breve (16 samples (23.5%)). The distribution of Bifidobacterium spp. was similar in the three feeding groups. In contrast, the high incidence of B. breve in cluster I, especially subcluster Ia and B. longum longum type in cluster II, especially subcluster IIa and IIc were characterized by T-RFLP method. Our results showed that the colonization of Bifidobacterium spp. in infant feces correlated with the T-RFLP clusters.
Collapse
Affiliation(s)
- Shinji Sakata
- Microbe Division/Japan Collection of Microorganisms, RIKEN BioResource Center, Wako, Saitama 351-0198, Japan.
| | | | | | | | | | | | | |
Collapse
|