101
|
Eid HM, Wright ML, Anil Kumar NV, Qawasmeh A, Hassan STS, Mocan A, Nabavi SM, Rastrelli L, Atanasov AG, Haddad PS. Significance of Microbiota in Obesity and Metabolic Diseases and the Modulatory Potential by Medicinal Plant and Food Ingredients. Front Pharmacol 2017; 8:387. [PMID: 28713266 PMCID: PMC5493053 DOI: 10.3389/fphar.2017.00387] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/02/2017] [Indexed: 01/11/2023] Open
Abstract
Metabolic syndrome is a cluster of three or more metabolic disorders including insulin resistance, obesity, and hyperlipidemia. Obesity has become the epidemic of the twenty-first century with more than 1.6 billion overweight adults. Due to the strong connection between obesity and type 2 diabetes, obesity has received wide attention with subsequent coining of the term "diabesity." Recent studies have identified unique contributions of the immensely diverse gut microbiota in the pathogenesis of obesity and diabetes. Several mechanisms have been proposed including altered glucose and fatty acid metabolism, hepatic fatty acid storage, and modulation of glucagon-like peptide (GLP)-1. Importantly, the relationship between unhealthy diet and a modified gut microbiota composition observed in diabetic or obese subjects has been recognized. Similarly, the role of diet rich in polyphenols and plant polysaccharides in modulating gut bacteria and its impact on diabetes and obesity have been the subject of investigation by several research groups. Gut microbiota are also responsible for the extensive metabolism of polyphenols thus modulating their biological activities. The aim of this review is to shed light on the composition of gut microbes, their health importance and how they can contribute to diseases as well as their modulation by polyphenols and polysaccharides to control obesity and diabetes. In addition, the role of microbiota in improving the oral bioavailability of polyphenols and hence in shaping their antidiabetic and antiobesity activities will be discussed.
Collapse
Affiliation(s)
- Hoda M. Eid
- Natural Health Products and Metabolic Diseases Laboratory, Department of Pharmacology and Physiology, Université de MontréalMontréal, QC, Canada
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic MedicinesMontréal, QC, Canada
- Department of Pharmacognosy, University of Beni-SuefBeni-Suef, Egypt
| | - Michelle L. Wright
- Nell Hodgson Woodruff School of Nursing, Emory UniversityAtlanta, GA, United States
| | - N. V. Anil Kumar
- Department of Chemistry, Manipal Institute of Technology, Manipal UniversityManipal, India
| | | | - Sherif T. S. Hassan
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences BrnoBrno, Czechia
| | - Andrei Mocan
- Department of Pharmaceutical Botany, Iuliu Hatieganu University of Medicine and PharmacyCluj-Napoca, Romania
- ICHAT and Institute for Life Sciences, University of Agricultural Sciences and Veterinary MedicineCluj-Napoca, Romania
| | - Seyed M. Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical SciencesTehran, Iran
| | - Luca Rastrelli
- Dipartimento di Farmacia, University of SalernoFisciano, Italy
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Breeding, Polish Academy of SciencesJastrzebiec, Poland
- Department of Pharmacognosy, University of ViennaVienna, Austria
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of ViennaVienna, Austria
| | - Pierre S. Haddad
- Natural Health Products and Metabolic Diseases Laboratory, Department of Pharmacology and Physiology, Université de MontréalMontréal, QC, Canada
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic MedicinesMontréal, QC, Canada
| |
Collapse
|
102
|
Variation between the oral and faecal microbiota in a free-living passerine bird, the great tit (Parus major). PLoS One 2017; 12:e0179945. [PMID: 28662106 PMCID: PMC5491070 DOI: 10.1371/journal.pone.0179945] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/07/2017] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract of vertebrates is inhabited by diverse bacterial communities that induce marked effects on the host physiology and health status. The composition of the gastrointestinal microbiota is characterized by pronounced taxonomic and functional variability among different regions of the vertebrate gastrointestinal tract. Despite the relatively solid knowledge on the among-region variations of the gastrointestinal microbiota in model mammalian species, there are only a few studies concerning among-region variations of the gastrointestinal microbiota in free-living non-mammalian vertebrate taxa. We used Illumina MiSeq sequencing of bacterial 16S rRNA amplicons to compare the diversity as well as taxonomic composition of bacterial communities in proximal vs. distal parts of the gastrointestinal tract (represented by oral swabs and faecal samples, respectively) in a wild passerine bird, the great tit (Parus major). The diversity of the oral microbiota was significantly higher compared to the faecal microbiota, whereas interindividual variation was higher in faecal than in oral samples. We also observed a pronounced difference in taxonomic content between the oral and faecal microbiota. Bacteria belonging to the phyla Proteobacteria, Firmicutes and Actinobacteria typically dominated in both oral and faecal samples. A high abundance of bacteria belonging to Tenericutes was observed only in faecal samples. Surprisingly, we found only a slight correlation between the faecal and oral microbiota at the within-individual level, suggesting that the microbial composition in these body sites is shaped by independent regulatory processes. Given the independence of these two communities at the individual level, we propose that simultaneous sampling of the faecal and oral microbiota will extend our understanding of host vs. microbiota interactions in wild populations.
Collapse
|
103
|
Contributions of the maternal oral and gut microbiome to placental microbial colonization in overweight and obese pregnant women. Sci Rep 2017; 7:2860. [PMID: 28588199 PMCID: PMC5460277 DOI: 10.1038/s41598-017-03066-4] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/21/2017] [Indexed: 01/01/2023] Open
Abstract
A distinct bacterial signature of the placenta was reported, providing evidence that the fetus does not develop in a sterile environment. The oral microbiome was suggested as a possible source of the bacterial DNA present in the placenta based on similarities to the oral non-pregnant microbiome. Here, the possible origin of the placental microbiome was assessed, examining the gut, oral and placental microbiomes from the same pregnant women. Microbiome profiles from 37 overweight and obese pregnant women were examined by 16SrRNA sequencing. Fecal and oral contributions to the establishment of the placental microbiome were evaluated. Core phylotypes between body sites and metagenome predictive functionality were determined. The placental microbiome showed a higher resemblance and phylogenetic proximity with the pregnant oral microbiome. However, similarity decreased at lower taxonomic levels and microbiomes clustered based on tissue origin. Core genera: Prevotella, Streptococcus and Veillonella were shared between all body compartments. Pathways encoding tryptophan, fatty-acid metabolism and benzoate degradation were highly enriched specifically in the placenta. Findings demonstrate that the placental microbiome exhibits a higher resemblance with the pregnant oral microbiome. Both oral and gut microbiomes contribute to the microbial seeding of the placenta, suggesting that placental colonization may have multiple niche sources.
Collapse
|
104
|
Gyuraszova M, Kovalcikova A, Gardlik R. Association between oxidative status and the composition of intestinal microbiota along the gastrointestinal tract. Med Hypotheses 2017; 103:81-85. [DOI: 10.1016/j.mehy.2017.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 04/19/2017] [Indexed: 02/08/2023]
|
105
|
Zhang Y, Simon SE, Johnson JA, Allen MS. Spatial Microbial Composition Along the Gastrointestinal Tract of Captive Attwater's Prairie Chicken. MICROBIAL ECOLOGY 2017; 73:966-977. [PMID: 27752719 DOI: 10.1007/s00248-016-0870-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/28/2016] [Indexed: 06/06/2023]
Abstract
Gastrointestinal microbiota is increasingly recognized as an important component of individual health, and therefore, our ability to quantify its diversity accurately is central for exploring different ways to improve health. Non-invasive sampling methods, such as cloaca swabs, are often used to measure gastrointestinal microbiota diversity within an individual. However, few studies have addressed to what degree differences exist in microbial community composition along the gastrointestinal tract, and measures obtained from the cloaca may not actually represent the diversity present elsewhere in the gastrointestinal tract. In this study, we systematically characterized the gastrointestinal microbial community of the critically endangered Attwater's Prairie chicken (Tympanuchus cupido attwateri) by opportunistically sampling four different locations (ileum, cecum, large intestine, and cloaca) along the gastrointestinal tract of eight individuals. Spatial variation of microbial community was observed at different sampling locations within the gastrointestinal tract. The cecum harbored the most diverse and significantly different microbiota from the other locations, while the microbial α- and β-diversities were similar in the ileum, large intestine, and cloaca. The results of this study provide evidence that microbiota diversity can differ depending on sampling location and metric used to quantify diversity. As shown here, non-invasive cloacal sampling strategies may reflect microbiota diversity elsewhere in the gastrointestinal tract, yet caution is warranted when making generalizations in terms of the microbiota diversity correlations when samples are obtained from a single location within the gastrointestinal tract.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA
- Center for Biosafety and Biosecurity, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA
| | - Stephanie E Simon
- Department of Biological Sciences, Institute of Applied Sciences, University of North Texas, 1155 Union Circle #310559, Denton, TX, 76203-5017, USA
| | - Jeff A Johnson
- Department of Biological Sciences, Institute of Applied Sciences, University of North Texas, 1155 Union Circle #310559, Denton, TX, 76203-5017, USA.
| | - Michael S Allen
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA.
- Center for Biosafety and Biosecurity, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107-2699, USA.
| |
Collapse
|
106
|
Tropini C, Earle KA, Huang KC, Sonnenburg JL. The Gut Microbiome: Connecting Spatial Organization to Function. Cell Host Microbe 2017; 21:433-442. [PMID: 28407481 DOI: 10.1016/j.chom.2017.03.010] [Citation(s) in RCA: 383] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The first rudimentary evidence that the human body harbors a microbiota hinted at the complexity of host-associated microbial ecosystems. Now, almost 400 years later, a renaissance in the study of microbiota spatial organization, driven by coincident revolutions in imaging and sequencing technologies, is revealing functional relationships between biogeography and health, particularly in the vertebrate gut. In this Review, we present our current understanding of principles governing the localization of intestinal bacteria, and spatial relationships between bacteria and their hosts. We further discuss important emerging directions that will enable progressing from the inherently descriptive nature of localization and -omics technologies to provide functional, quantitative, and mechanistic insight into this complex ecosystem.
Collapse
Affiliation(s)
- Carolina Tropini
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kristen A Earle
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kerwyn Casey Huang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | | |
Collapse
|
107
|
Complete Genome Sequence of the Gamma-Aminobutyric Acid-Producing Strain Streptococcus thermophilus APC151. GENOME ANNOUNCEMENTS 2017; 5:5/17/e00205-17. [PMID: 28450504 PMCID: PMC5408102 DOI: 10.1128/genomea.00205-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Here is presented the whole-genome sequence of Streptococcus thermophilus APC151, isolated from a marine fish. This bacterium produces gamma-aminobutyric acid (GABA) in high yields and is biotechnologically suitable to produce naturally GABA-enriched biofunctional yogurt. Its complete genome comprises 2,097 genes and 1,839,134 nucleotides, with an average G+C content of 39.1%.
Collapse
|
108
|
Zaheer R, Dugat-Bony E, Holman D, Cousteix E, Xu Y, Munns K, Selinger LJ, Barbieri R, Alexander T, McAllister TA, Selinger LB. Changes in bacterial community composition of Escherichia coli O157:H7 super-shedder cattle occur in the lower intestine. PLoS One 2017; 12:e0170050. [PMID: 28141846 PMCID: PMC5283656 DOI: 10.1371/journal.pone.0170050] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 12/28/2016] [Indexed: 12/13/2022] Open
Abstract
Escherichia coli O157:H7 is a foodborne pathogen that colonizes ruminants. Cattle are considered the primary reservoir of E. coli O157:H7 with super-shedders, defined as individuals excreting > 104E. coli O157:H7 CFU g-1 feces. The mechanisms leading to the super-shedding condition are largely unknown. Here, we used 16S rRNA gene pyrosequencing to examine the composition of the fecal bacterial community in order to investigate changes in the bacterial microbiota at several locations along the digestive tract (from the duodenum to the rectal-anal junction) in 5 steers previously identified as super-shedders and 5 non-shedders. The overall bacterial community structure did not differ by E. coli O157:H7 shedding status; but several differences in the relative abundance of taxa and OTUs were noted between the two groups. The genus Prevotella was most enriched in the non-shedders while the genus Ruminococcus and the Bacteroidetes phylum were notably enriched in the super-shedders. There was greater bacterial diversity and richness in samples collected from the lower- as compared to the upper gastrointestinal tract (GI). The spiral colon was the only GI location that differed in terms of bacterial diversity between super-shedders and non-shedders. These findings reinforced linkages between E. coli O157:H7 colonization in cattle and the nature of the microbial community inhabiting the digestive tract of super-shedders.
Collapse
Affiliation(s)
- Rahat Zaheer
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Eric Dugat-Bony
- UMR Génie et Microbiologie des Procédés Alimentaires, AgroParisTech, INRA, Université Paris-Saclay, Thiverval-Grignon, France
| | - Devon Holman
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Elodie Cousteix
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Yong Xu
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Krysty Munns
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Lorna J Selinger
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Rutn Barbieri
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Trevor Alexander
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Tim A McAllister
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - L Brent Selinger
- Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada
| |
Collapse
|
109
|
Patrascu O, Béguet-Crespel F, Marinelli L, Le Chatelier E, Abraham AL, Leclerc M, Klopp C, Terrapon N, Henrissat B, Blottière HM, Doré J, Béra-Maillet C. A fibrolytic potential in the human ileum mucosal microbiota revealed by functional metagenomic. Sci Rep 2017; 7:40248. [PMID: 28091525 PMCID: PMC5238381 DOI: 10.1038/srep40248] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/05/2016] [Indexed: 12/26/2022] Open
Abstract
The digestion of dietary fibers is a major function of the human intestinal microbiota. So far this function has been attributed to the microorganisms inhabiting the colon, and many studies have focused on this distal part of the gastrointestinal tract using easily accessible fecal material. However, microbial fermentations, supported by the presence of short-chain fatty acids, are suspected to occur in the upper small intestine, particularly in the ileum. Using a fosmid library from the human ileal mucosa, we screened 20,000 clones for their activities against carboxymethylcellulose and xylans chosen as models of the major plant cell wall (PCW) polysaccharides from dietary fibres. Eleven positive clones revealed a broad range of CAZyme encoding genes from Bacteroides and Clostridiales species, as well as Polysaccharide Utilization Loci (PULs). The functional glycoside hydrolase genes were identified, and oligosaccharide break-down products examined from different polysaccharides including mixed-linkage β-glucans. CAZymes and PULs were also examined for their prevalence in human gut microbiome. Several clusters of genes of low prevalence in fecal microbiome suggested they belong to unidentified strains rather specifically established upstream the colon, in the ileum. Thus, the ileal mucosa-associated microbiota encompasses the enzymatic potential for PCW polysaccharide degradation in the small intestine.
Collapse
Affiliation(s)
- Orlane Patrascu
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Fabienne Béguet-Crespel
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Ludovica Marinelli
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | | | - Anne-Laure Abraham
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Marion Leclerc
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Christophe Klopp
- Plate-forme bio-informatique Genotoul, Mathématiques et Informatique Appliquées de Toulouse, INRA, Castanet-Tolosan, France
| | - Nicolas Terrapon
- CNRS UMR 7257, Université Aix-Marseille, 13288 Marseille, France.,INRA, USC 1408 AFMB, 13288 Marseille, France
| | - Bernard Henrissat
- CNRS UMR 7257, Université Aix-Marseille, 13288 Marseille, France.,INRA, USC 1408 AFMB, 13288 Marseille, France.,Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hervé M Blottière
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.,Metagenopolis, INRA, 78350 Jouy-en-Josas, France
| | - Joël Doré
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.,Metagenopolis, INRA, 78350 Jouy-en-Josas, France
| | - Christel Béra-Maillet
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| |
Collapse
|
110
|
Changes in duodenal tissue-associated microbiota following hookworm infection and consecutive gluten challenges in humans with coeliac disease. Sci Rep 2016; 6:36797. [PMID: 27827438 PMCID: PMC5101533 DOI: 10.1038/srep36797] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/21/2016] [Indexed: 12/26/2022] Open
Abstract
A reduced diversity of the gastrointestinal commensal microbiota is associated with the development of several inflammatory diseases. Recent reports in humans and animal models have demonstrated the beneficial therapeutic effects of infections by parasitic worms (helminths) in some inflammatory disorders, such as inflammatory bowel disease (IBD) and coeliac disease (CeD). Interestingly, these studies have described how helminths may alter the intestinal microbiota, potentially representing a mechanism by which they regulate inflammation. However, for practical reasons, these reports have primarily analysed the faecal microbiota. In the present investigation, we have assessed, for the first time, the changes in the microbiota at the site of infection by a parasitic helminth (hookworm) and gluten-dependent inflammation in humans with CeD using biopsy tissue from the duodenum. Hookworm infection and gluten exposure were associated with an increased abundance of species
within the Bacteroides phylum, as well as increases in the richness and diversity of the tissue-resident microbiota within the intestine, results that are consistent with previous reports using other helminth species in humans and animal models. Hence, this may represent a mechanism by which parasitic helminths may restore intestinal immune homeostasis and exert a therapeutic benefit in CeD, and potentially other inflammatory disorders.
Collapse
|
111
|
Abstract
The relevance of biogeography to the distal gut microbiota has been investigated in both health and inflammatory bowel disease (IBD), however multiple factors, including sample type and methodology, microbiota characterization and interpersonal variability make the construction of a core model of colonic biogeography challenging. In addition, how phylogenetic classification relates to immunogenicity and whether consistent alterations in the microbiota are associated with ulcerative colitis (UC) remain open questions. This addendum seeks to review the human colonic microbiota in health and UC as currently understood, in the broader context of the human microbiome.
Collapse
Affiliation(s)
- Aonghus Lavelle
- School of Medicine and Medical Science,
University College Dublin, Dublin, Ireland,Centre for Colorectal Disease, St.
Vincent's University Hospital, Dublin,
Ireland
| | - Grainne Lennon
- School of Medicine and Medical Science,
University College Dublin, Dublin, Ireland,Centre for Colorectal Disease, St.
Vincent's University Hospital, Dublin,
Ireland
| | - Desmond C. Winter
- School of Medicine and Medical Science,
University College Dublin, Dublin, Ireland,Centre for Colorectal Disease, St.
Vincent's University Hospital, Dublin,
Ireland
| | - P. Ronan O'Connell
- School of Medicine and Medical Science,
University College Dublin, Dublin, Ireland,Centre for Colorectal Disease, St.
Vincent's University Hospital, Dublin,
Ireland
| |
Collapse
|
112
|
Distal, not proximal, colonic acetate infusions promote fat oxidation and improve metabolic markers in overweight/obese men. Clin Sci (Lond) 2016; 130:2073-2082. [PMID: 27439969 DOI: 10.1042/cs20160263] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/20/2016] [Indexed: 12/25/2022]
Abstract
Gut microbial-derived short-chain fatty acids (SCFA) are believed to affect host metabolism and cardiometabolic risk factors. The present study aim was to investigate the effects of proximal and distal colonic infusions with the SCFA acetate on fat oxidation and other metabolic parameters in men. In this randomized, double-blind crossover trial, six overweight/obese men [body mass index (BMI) 25-35 kg/m2] underwent two experimental periods: one with distal and one with proximal colonic sodium acetate infusions. A feeding catheter was endoscopically positioned at the beginning of each period and remained in the colon for three consecutive test days, enabling colonic acetate (100 or 180 mmol/l) or placebo infusion during fasting conditions and after an oral glucose load (postprandial). Fat oxidation and energy expenditure were measured using an open-circuit ventilated hood system and blood samples were repeatedly collected for 2 h during fasting and postprandial conditions. Distal colonic 180 mmol/l acetate infusions increased fasting fat oxidation (1.78±0.28 compared with -0.78±0.89 g fat 2 h-1, P=0.015), fasting peptide YY (PYY, P=0.01) and postprandial glucose and insulin concentrations (P<0.05), and tended to increase fasting plasma acetate (P=0.069) compared with placebo. Distal 100 mmol/l acetate administration tended to decrease fasting tumour necrosis factor-α (TNF-α; P=0.067) compared with placebo. In contrast, proximal colonic acetate infusions showed no effects on substrate metabolism, circulating hormones or inflammatory markers. In conclusion distal colonic acetate infusions affected whole-body substrate metabolism, with a pronounced increase in fasting fat oxidation and plasma PYY. Modulating colonic acetate may be a nutritional target to treat or prevent metabolic disorders.
Collapse
|
113
|
Vindigni SM, Zisman TL, Suskind DL, Damman CJ. The intestinal microbiome, barrier function, and immune system in inflammatory bowel disease: a tripartite pathophysiological circuit with implications for new therapeutic directions. Therap Adv Gastroenterol 2016; 9:606-25. [PMID: 27366227 PMCID: PMC4913337 DOI: 10.1177/1756283x16644242] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We discuss the tripartite pathophysiological circuit of inflammatory bowel disease (IBD), involving the intestinal microbiota, barrier function, and immune system. Dysfunction in each of these physiological components (dysbiosis, leaky gut, and inflammation) contributes in a mutually interdependent manner to IBD onset and exacerbation. Genetic and environmental risk factors lead to disruption of gut homeostasis: genetic risks predominantly affect the immune system, environmental risks predominantly affect the microbiota, and both affect barrier function. Multiple genetic and environmental 'hits' are likely necessary to establish and exacerbate disease. Most conventional IBD therapies currently target only one component of the pathophysiological circuit, inflammation; however, many patients with IBD do not respond to immune-modulating therapies. Hope lies in new classes of therapies that target the microbiota and barrier function.
Collapse
Affiliation(s)
- Stephen M. Vindigni
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Timothy L. Zisman
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - David L. Suskind
- Department of Pediatrics, Seattle Children’s Hospital and University of Washington, Seattle, WA, USA
| | | |
Collapse
|
114
|
Gupta RS. Impact of genomics on the understanding of microbial evolution and classification: the importance of Darwin's views on classification. FEMS Microbiol Rev 2016; 40:520-53. [PMID: 27279642 DOI: 10.1093/femsre/fuw011] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2016] [Indexed: 12/24/2022] Open
Abstract
Analyses of genome sequences, by some approaches, suggest that the widespread occurrence of horizontal gene transfers (HGTs) in prokaryotes disguises their evolutionary relationships and have led to questioning of the Darwinian model of evolution for prokaryotes. These inferences are critically examined in the light of comparative genome analysis, characteristic synapomorphies, phylogenetic trees and Darwin's views on examining evolutionary relationships. Genome sequences are enabling discovery of numerous molecular markers (synapomorphies) such as conserved signature indels (CSIs) and conserved signature proteins (CSPs), which are distinctive characteristics of different prokaryotic taxa. Based on these molecular markers, exhibiting high degree of specificity and predictive ability, numerous prokaryotic taxa of different ranks, currently identified based on the 16S rRNA gene trees, can now be reliably demarcated in molecular terms. Within all studied groups, multiple CSIs and CSPs have been identified for successive nested clades providing reliable information regarding their hierarchical relationships and these inferences are not affected by HGTs. These results strongly support Darwin's views on evolution and classification and supplement the current phylogenetic framework based on 16S rRNA in important respects. The identified molecular markers provide important means for developing novel diagnostics, therapeutics and for functional studies providing important insights regarding prokaryotic taxa.
Collapse
Affiliation(s)
- Radhey S Gupta
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
115
|
Boland M. Human digestion--a processing perspective. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2016; 96:2275-2283. [PMID: 26711173 DOI: 10.1002/jsfa.7601] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 11/14/2015] [Accepted: 12/20/2015] [Indexed: 06/05/2023]
Abstract
The human digestive system is reviewed in the context of a process with four major unit operations: oral processing to reduce particle size and produce a bolus; gastric processing to initiate chemical and enzymatic breakdown; small intestinal processing to break down macromolecules and absorb nutrients; and fermentation and water removal in the colon. Topics are highlighted about which we need to know more, including effects of aging and dentition on particle size in the bolus, effects of different patterns of food and beverage intake on nutrition, changes in saliva production and composition, mechanical effects of gastric processing, distribution of pH in the stomach, physicochemical and enzymatic effects on nutrient availability and uptake in the small intestine, and the composition, effects of and changes in the microbiota of the colon. Current topics of interest including food synergy, gut-brain interactions, nutritional phenotype and digestion in the elderly are considered. Finally, opportunities for food design based on an understanding of digestive processing are discussed.
Collapse
Affiliation(s)
- Mike Boland
- Riddet Institute, Palmerston North, New Zealand
| |
Collapse
|
116
|
Chen J, Xie P, Lin J, He J, Zeng C, Chen J. Effects of microcystin-LR on gut microflora in different gut regions of mice. J Toxicol Sci 2016; 40:485-94. [PMID: 26165645 DOI: 10.2131/jts.40.485] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
To reveal the toxicological effects of the hepatotoxic microcystin-leucine arginine (MC-LR) on gut microbial community composition in different gut regions, we conducted a subchronic exposure of BALB/c mice to MC-LR via intragastric administration. Denaturing gradient gel electrophoresis (DGGE) was employed to profile the shifts of microbes after MC-LR treatment in the jejuno-ileum, caecum and colon. DGGE profiles analysis showed that MC-LR increased the microbial species richness (number of microbial bands) in the caecum and colon as well as microbial diversity (Shannon-Wiener index) in the caecum. The cluster analysis of DGGE profiles indicated that the microbial structures in the caecum and colon shifted significantly after MC-LR treatment, while that in the jejuno-ileum did not. All the relatively decreased gut microbes belonged to Clostridia in the Firmicutes phylum, and most of them were Lachnospiraceae. The increased ones derived from a variety of microbes including species from Porphyromonadaceae and Prevotellaceae in the Bacteroidetes phylum, as well as Lachnospiraceae and Ruminococcaceae in the Firmicutes phylum, and among which, the increase of Barnesiella in Porphyromonadaceae was most remarkable. In conclusion, subchronic exposure to MC-LR could disturb the balance of gut microbes in mice, and its toxicological effects varied between the jejuno-ileum and the other two gut regions.
Collapse
Affiliation(s)
- Jing Chen
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, China
| | | | | | | | | | | |
Collapse
|
117
|
Zadeh-Tahmasebi M, Duca FA, Rasmussen BA, Bauer PV, Côté CD, Filippi BM, Lam TKT. Activation of Short and Long Chain Fatty Acid Sensing Machinery in the Ileum Lowers Glucose Production in Vivo. J Biol Chem 2016; 291:8816-24. [PMID: 26896795 DOI: 10.1074/jbc.m116.718460] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Indexed: 01/14/2023] Open
Abstract
Evidence continues to emerge detailing the myriad of ways the gut microbiota influences host energy homeostasis. Among the potential mechanisms, short chain fatty acids (SCFAs), the byproducts of microbial fermentation of dietary fibers, exhibit correlative beneficial metabolic effects in humans and rodents, including improvements in glucose homeostasis. The underlying mechanisms, however, remain elusive. We here report that one of the main bacterially produced SCFAs, propionate, activates ileal mucosal free fatty acid receptor 2 to trigger a negative feedback pathway to lower hepatic glucose production in healthy rats in vivo We further demonstrate that an ileal glucagon-like peptide-1 receptor-dependent neuronal network is necessary for ileal propionate and long chain fatty acid sensing to regulate glucose homeostasis. These findings highlight the potential to manipulate fatty acid sensing machinery in the ileum to regulate glucose homeostasis.
Collapse
Affiliation(s)
- Melika Zadeh-Tahmasebi
- From the Toronto General Research Institute and Department of Medicine, UHN, Toronto, Ontario M5G 1L7, the Departments of Physiology and
| | - Frank A Duca
- From the Toronto General Research Institute and Department of Medicine, UHN, Toronto, Ontario M5G 1L7
| | - Brittany A Rasmussen
- From the Toronto General Research Institute and Department of Medicine, UHN, Toronto, Ontario M5G 1L7, the Departments of Physiology and
| | - Paige V Bauer
- From the Toronto General Research Institute and Department of Medicine, UHN, Toronto, Ontario M5G 1L7, the Departments of Physiology and
| | - Clémence D Côté
- From the Toronto General Research Institute and Department of Medicine, UHN, Toronto, Ontario M5G 1L7, the Departments of Physiology and
| | - Beatrice M Filippi
- From the Toronto General Research Institute and Department of Medicine, UHN, Toronto, Ontario M5G 1L7
| | - Tony K T Lam
- From the Toronto General Research Institute and Department of Medicine, UHN, Toronto, Ontario M5G 1L7, the Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario M5S 1A8, and the Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
118
|
Complete Genome Sequence of Streptococcus salivarius HSISS4, a Human Commensal Bacterium Highly Prevalent in the Digestive Tract. GENOME ANNOUNCEMENTS 2016; 4:4/1/e01637-15. [PMID: 26847886 PMCID: PMC4742683 DOI: 10.1128/genomea.01637-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The human commensal bacterium Streptococcus salivarius plays a major role in the equilibrium of microbial communities of the digestive tract. Here, we report the first complete genome sequence of a Streptococcus salivarius strain isolated from the small intestine, namely, HSISS4. Its circular chromosome comprises 1,903 coding sequences and 2,100,988 nucleotides.
Collapse
|
119
|
Qin HP, Wang XH. Relationship between small intestinal microbiota and bowel and metabolic diseases. Shijie Huaren Xiaohua Zazhi 2016; 24:375-381. [DOI: 10.11569/wcjd.v24.i3.375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Microbiota plays a vital role in human health and diseases. Colonic microbiota has been deeply studied because it is abundant and easy to get. The small intestine is the main place where most nutrients are digested and absorbed, and the microbiota which dwells in the small intestine has also profound effects on the host. As it is difficult to obtain samples from the small intestine, small intestinal microbiota composition is seldom reported. A few recent studies show that a significant distinction exists in microbiota between the small intestine and colon. The small intestinal microbiota participates in energy storage, intestinal endocrine function and immune maturation of the host. Therefore, more and more studies are focusing on the small intestinal microbiota. This paper reviews recent advances in understanding the relationship between small intestinal microbiota and related diseases.
Collapse
|
120
|
Stability and Inhibitory Activity of Pediocin PA-1 Against Listeria sp. in Simulated Physiological Conditions of the Human Terminal Ileum. Probiotics Antimicrob Proteins 2016; 4:250-8. [PMID: 26782185 DOI: 10.1007/s12602-012-9111-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Listeria monocytogenes is responsible for severe foodborne infections, which can be life-threatening especially for infants and elderly populations. The emergence of antibiotic-resistant pathogens has stimulated the search for new strategies, such as the use of bacteriocins, to prevent or cure foodborne infectious diseases in the intestine. In this study, we evaluated the efficacy of the bacteriocin pediocin PA-1 from Pediococcus acidilactici UL5 to inhibit Listeria ivanovii, used as a surrogate for L. monocytogenes, under physiological conditions of the terminal ileum, simulated in a continuous in vitro fermentation model. A fecal sample from a healthy adult was immobilized and propagated for 30 days in a continuous stirred tank reactor, fed with a nutritive medium simulating the ileal chime (pH 7.5). After reaching a pseudo-steady state, the reactor was inoculated five times with L. ivanovii to reach a final concentration of 10(7) CFU/ml within the reactor. Two spikes of L. ivanovii without adjunction of pediocin PA-1 served as control assays, and three other spikes were done to test the effects of three concentrations of pediocin PA-1 corresponding to 2, 3, and 5× the minimum inhibitory concentration (MIC) active against L. ivanovii. The concentration of L. ivanovii in the reactor was followed for 8 h using the PALCAM selective medium. The different groups of commensal bacteria were enumerated on selective medium or using fluorescence in situ hybridization. Our data showed that pediocin PA-1 is stable in the ileum conditions and that it is able to exert its inhibition activity against L. ivanovii in a dose-dependent manner. The addition of pediocin PA-1 at 5 × MIC induced a complete disappearance of L. ivanovii (5 log reduction) within 5 h, compared to a reduction of 2 logs, corresponding to the washout phenomenon, when no pediocin PA-1 was added. Reduction of 0.8 and 1.3 logs within 8 h was also obtained with the addition of 2 and 3 × MIC, respectively. The same experiment has shown that addition of pediocin-PA1 in the reactor had a negligible effect on the balance of commensal bacteria.
Collapse
|
121
|
Fuhler GM, Parikh K, van der Woude CJ, Peppelenbosch MP. Linkage between genotype and immunological phenotype in Crohn's disease. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:237. [PMID: 26539454 DOI: 10.3978/j.issn.2305-5839.2015.09.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Understanding the mechanisms that drive uncontrolled inflammation in Crohn's disease (CD) remains one of the most pressing challenges in contemporary experimental medicine. Recently, a three-phased view on the pathogenesis of CD was proposed in which following the breakdown of intestinal epithelial barrier function, CD patients fail to clear the resulting infectious debris, provoking subsequent immune responses. This view on CD is attractive in that it is testable and allows better diagnosis of disease if proven correct, apart from opening a window on new therapeutic horizons. Here we shall argue, however, that this scheme may be an oversimplification in that it ignores the genetic diversity of CD and thus does not fully take into account the nature of the intestinal epithelium, which appears a non-passive actor in this disease.
Collapse
Affiliation(s)
- Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Erasmus University of Rotterdam, Rotterdam, The Netherlands
| | - Kaushal Parikh
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Erasmus University of Rotterdam, Rotterdam, The Netherlands
| | - C Janneke van der Woude
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Erasmus University of Rotterdam, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Erasmus University of Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
122
|
First report on the bacterial diversity in the distal gut of dholes (Cuon alpinus) by using 16S rRNA gene sequences analysis. J Appl Genet 2015; 57:275-83. [PMID: 26423781 DOI: 10.1007/s13353-015-0319-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/19/2015] [Accepted: 09/14/2015] [Indexed: 12/15/2022]
Abstract
The aim of this study was to investigate the bacterial community in the distal gut of dholes (Cuon alpinus) based on the analysis of bacterial 16S rRNA gene sequences. Fecal samples were collected from five healthy unrelated dholes captured from Qilian Mountain in Gansu province of China. The diversity of the fecal bacteria community was investigated by constructing a polymerase chain reaction (PCR)-amplified 16S rRNA gene clone library. Bacterial 16S rRNA gene was amplified by using universal bacterial primers 27F and 1492R. A total of 275 chimera-free near full length 16S rRNA gene sequences were collected, and 78 non-redundant bacteria phylotypes (operational taxonomical units, OTUs) were identified according to the 97 % sequence similarity. Forty-two OTUs (53.8 %) showed less than 98 % sequence similarity to 16S rRNA gene sequences reported previously. Phylogenetic analysis demonstrated that dhole bacterial community comprised five different phyla, with the majority of sequences being classified within the phylum Bacteroidetes (64.7 %), followed by Firmicutes (29.8 %), Fusobacteria (4.7 %),Proteobacteria (0.4 %), and Actinobacteria (0.4 %). The only order Bacteroidales in phylum Bacteroidetes was the most abundant bacterial group in the intestinal bacterial community of dholes. Firmicutes and Bacteroidetes were the two most diverse bacterial phyla with 46.2 and 44.9 % of OTUs contained, respectively. Bacteroidales and Clostridiales were the two most diverse bacterial orders that contained 44.9 and 39.7 % of OTUs, respectively.
Collapse
|
123
|
Angelakis E, Armougom F, Carrière F, Bachar D, Laugier R, Lagier JC, Robert C, Michelle C, Henrissat B, Raoult D. A Metagenomic Investigation of the Duodenal Microbiota Reveals Links with Obesity. PLoS One 2015; 10:e0137784. [PMID: 26356733 PMCID: PMC4565581 DOI: 10.1371/journal.pone.0137784] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/28/2015] [Indexed: 01/16/2023] Open
Abstract
Background Few studies have tested the small intestine microbiota in humans, where most nutrient digestion and absorption occur. Here, our objective was to examine the duodenal microbiota between obese and normal volunteers using metagenomic techniques. Methodology/Principal Findings We tested duodenal samples from five obese and five normal volunteers using 16S rDNA V6 pyrosequencing and Illumina MiSeq deep sequencing. The predominant phyla of the duodenal microbiota were Firmicutes and Actinobacteria, whereas Bacteroidetes were absent. Obese individuals had a significant increase in anaerobic genera (p < 0.001) and a higher abundance of genes encoding Acyl-CoA dehydrogenase (p = 0.0018) compared to the control group. Obese individuals also had a reduced abundance of genes encoding sucrose phosphorylase (p = 0.015) and 1,4-alpha-glucan branching enzyme (p = 0.05). Normal weight people had significantly increased FabK (p = 0.027), and the glycerophospholipid metabolism pathway revealed the presence of phospholipase A1 only in the control group (p = 0.05). Conclusions/Significance The duodenal microbiota of obese individuals exhibit alterations in the fatty acid and sucrose breakdown pathways, probably induced by diet imbalance.
Collapse
Affiliation(s)
- Emmanouil Angelakis
- URMITE CNRS-IRD 198 UMR 6236, Aix Marseille Université, Faculté de Médecine, 27 Bd Jean Moulin, 13385, Marseille, France
| | - Fabrice Armougom
- URMITE CNRS-IRD 198 UMR 6236, Aix Marseille Université, Faculté de Médecine, 27 Bd Jean Moulin, 13385, Marseille, France
| | - Frédéric Carrière
- CNRS, Aix Marseille Université, UMR7282 Enzymology at Interfaces and Physiology of Lipolysis, 13009, Marseille, France
| | - Dipankar Bachar
- URMITE CNRS-IRD 198 UMR 6236, Aix Marseille Université, Faculté de Médecine, 27 Bd Jean Moulin, 13385, Marseille, France
| | - René Laugier
- Hepato-gastroenterology Department, Hôpital de la Timone, Marseille, France
| | - Jean-Christophe Lagier
- URMITE CNRS-IRD 198 UMR 6236, Aix Marseille Université, Faculté de Médecine, 27 Bd Jean Moulin, 13385, Marseille, France
| | - Catherine Robert
- URMITE CNRS-IRD 198 UMR 6236, Aix Marseille Université, Faculté de Médecine, 27 Bd Jean Moulin, 13385, Marseille, France
| | - Caroline Michelle
- URMITE CNRS-IRD 198 UMR 6236, Aix Marseille Université, Faculté de Médecine, 27 Bd Jean Moulin, 13385, Marseille, France
| | - Bernard Henrissat
- Architecture et Fonction des Macromolécules Biologiques, Centre National de la Recherche Scientifique, Aix-Marseille Université, 13288, Marseille, France
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Didier Raoult
- URMITE CNRS-IRD 198 UMR 6236, Aix Marseille Université, Faculté de Médecine, 27 Bd Jean Moulin, 13385, Marseille, France
- * E-mail:
| |
Collapse
|
124
|
Derrien M, van Hylckama Vlieg JE. Fate, activity, and impact of ingested bacteria within the human gut microbiota. Trends Microbiol 2015; 23:354-66. [DOI: 10.1016/j.tim.2015.03.002] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/28/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023]
|
125
|
Composition and diversity of the bacterial community in snow leopard (Uncia uncia) distal gut. ANN MICROBIOL 2015. [DOI: 10.1007/s13213-014-0909-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
126
|
Kim DH. Gut Microbiota-Mediated Drug-Antibiotic Interactions. Drug Metab Dispos 2015; 43:1581-9. [PMID: 25926432 DOI: 10.1124/dmd.115.063867] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/29/2015] [Indexed: 12/19/2022] Open
Abstract
Xenobiotic metabolism involves the biochemical modification of drugs and phytochemicals in living organisms, including humans and other animals. In the intestine, the gut microbiota catalyzes the conversion of hydrophilic drugs into absorbable, hydrophobic compounds through hydroxyzation and reduction. Drugs and phytochemicals are transformed into bioactive (sulfasalazine, lovastatin, and ginsenoside Rb1), bioinactive (chloramphenicol, ranitidine, and metronidazole), and toxic metabolites (nitrazepam), thus affecting the pharmacokinetics of the original compounds. Antibiotics suppress the activities of drug-metabolizing enzymes by inhibiting the proliferation of gut microbiota. Antibiotic treatment might influence xenobiotic metabolisms more extensively and potently than previously recognized and reduce gut microbiota-mediated transformation of orally administered drugs, thereby altering the systemic concentrations of intact drugs, their metabolites, or both. This review describes the effects of antibiotics on the metabolism of drugs and phytochemicals by the gut microbiota.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
127
|
Anders H, Power JF, MacKenzie AD, Lagutin K, Vyssotski M, Hanssen E, Moreau JW, Stott MB. Limisphaera ngatamarikiensis gen. nov., sp. nov., a thermophilic, pink-pigmented coccus isolated from subaqueous mud of a geothermal hotspring. Int J Syst Evol Microbiol 2015; 65:1114-1121. [DOI: 10.1099/ijs.0.000063] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A novel bacterial strain, NGM72.4T, was isolated from a hot spring in the Ngatamariki geothermal field, New Zealand. Phylogenetic analysis based on 16S rRNA gene sequences grouped it into the phylum
Verrucomicrobia
and class level group 3 (also known as OPB35 soil group). NGM72.4T stained Gram-negative, and was catalase- and oxidase-positive. Cells were small cocci, 0.5–0.8 µm in diameter, which were motile by means of single flagella. Transmission electron micrograph (TEM) imaging showed an unusual pirellulosome-like intracytoplasmic membrane. The peptidoglycan content was very small with only trace levels of diaminopimelic acid detected. No peptidoglycan structure was visible in TEM imaging. The predominant isoprenoid quinone was MK-7 (92 %). The major fatty acids (>15 %) were C16 : 0, anteiso-C15 : 0, iso-C16 : 0 and anteiso-C17 : 0. Major phospholipids were phosphatidylethanolamine (PE), phosphatidylmonomethylethanolamine (PMME) and cardiolipin (CL), and a novel analogous series of phospholipids where diacylglycerol was replaced with diacylserinol (sPE, sPMME, sCL). The DNA G+C content was 65.6 mol%. Cells displayed an oxidative chemoheterotrophic metabolism. NGM72.4T is a strictly aerobic thermophile (growth optimum 60–65 °C), has a slightly alkaliphilic pH growth optimum (optimum pH 8.1–8.4) and has a NaCl tolerance of up to 8 g l−1. Colonies were small, circular and pigmented pale pink. The distinct phylogenetic position and phenotypic traits of strain NGM72.4T distinguish it from all other described species of the phylum
Verrucomicrobia
and, therefore, it is considered to represent a novel species in a new genus for which we propose the name Limisphaera ngatamarikiensis gen. nov., sp. nov. The type strain is NGM72.4T ( = ICMP 20182T = DSM 27329T).
Collapse
Affiliation(s)
- Heike Anders
- Lehrstuhl für Tierhygiene, Technische Universität München, Weihenstephaner Berg 3 0D-85354, Freising, Germany
- GNS Science, Extremophile Research Group, Private Bag 2000, Taupō 3352, New Zealand
| | - Jean F. Power
- GNS Science, Extremophile Research Group, Private Bag 2000, Taupō 3352, New Zealand
| | | | - Kirill Lagutin
- Callaghan Innovation, PO Box 31310, Lower Hutt 5040, New Zealand
| | | | - Eric Hanssen
- University of Melbourne, 30 Flemington Road, Victoria 3010, Australia
| | - John W. Moreau
- University of Melbourne, 30 Flemington Road, Victoria 3010, Australia
| | - Matthew B. Stott
- GNS Science, Extremophile Research Group, Private Bag 2000, Taupō 3352, New Zealand
| |
Collapse
|
128
|
García-Mazcorro JF, Garza-González E, Marroquín-Cardona AG, Tamayo JL. [Characterization, influence and manipulation of the gastrointestinal microbiota in health and disease]. GASTROENTEROLOGIA Y HEPATOLOGIA 2015; 38:445-66. [PMID: 25769877 DOI: 10.1016/j.gastrohep.2015.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 01/12/2015] [Accepted: 01/26/2015] [Indexed: 01/08/2023]
Abstract
The gastrointestinal tract harbors trillions of microorganisms that are indispensable for health. The gastrointestinal microbiota can be studied using culture and molecular methods. The applications of massive sequencing are constantly increasing, due to their high yield, increasingly accessible costs, and the availability of free software for data analysis. The present article provides a detailed review of a large number of studies on the gastrointestinal microbiota and its influence on human health; particular emphasis is placed on the evidence suggesting a relationship between the gastrointestinal microbial ecosystem and diverse physiological and immune/inflammatory processes. Discussion of the articles analyzed combines a medical approach and current concepts of microbial molecular ecology. The present revision aims to be useful to those interested in the gastrointestinal microbiota and its possible alteration to maintain, re-establish and enhance health in the human host.
Collapse
Affiliation(s)
- José F García-Mazcorro
- Facultad de Medicina Veterinaria, Universidad Autónoma de Nuevo León, General Escobedo, Nuevo León, México; Grupo de investigación Ecobiología Médica, Facultad de Medicina Veterinaria, Universidad Autónoma de Nuevo León, General Escobedo, Nuevo León, México.
| | - Elvira Garza-González
- Servicio de Gastroenterología y Departamento de Patología Clínica, Hospital Universitario «Dr. José Eleuterio González», Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Alicia G Marroquín-Cardona
- Facultad de Medicina Veterinaria, Universidad Autónoma de Nuevo León, General Escobedo, Nuevo León, México; Grupo de investigación Ecobiología Médica, Facultad de Medicina Veterinaria, Universidad Autónoma de Nuevo León, General Escobedo, Nuevo León, México; Departamento de Fisiología, Farmacología y Toxicología, Facultad de Medicina Veterinaria, Universidad Autónoma de Nuevo León, General Escobedo, Nuevo León, México
| | - José L Tamayo
- Centro de Investigación y Docencia en Ciencias de la Salud, Hospital Civil de Culiacán, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, México
| |
Collapse
|
129
|
Murimonas intestini gen. nov., sp. nov., an acetate-producing bacterium of the family Lachnospiraceae isolated from the mouse gut. Int J Syst Evol Microbiol 2015; 65:870-878. [DOI: 10.1099/ijs.0.000030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Three strains of an anaerobic, Gram-stain-positive coccobacillus were isolated from the intestines of mice. These strains shared 100 % similarity in their 16S rRNA gene sequences, but were distantly related to any described members of the family
Lachnospiraceae
(<94 %). The most closely related species with names that have standing in nomenclature were
Robinsoniella peoriensis
,
Ruminococcus gnavus
,
Blautia producta
and
Clostridium xylanolyticum
. Phylogenetic relationships based on 16S rRNA gene sequence analysis were confirmed by partial sequencing of hsp60 genes. The use of an in-house database search pipeline revealed that the new isolates are most prevalent in bovine gut samples when compared with human and mouse samples for
Ruminococcus gnavus
and
B. producta
. All three isolated strains shared similar cellular fatty acid patterns dominated by C16 : 0 methyl ester. Differences in the proportions of C12 : 0 methyl ester, C14 : 0 methyl ester and C18 : 1
cis-11 dimethyl acetal were observed when compared with phylogenetically neighbouring species. The major short-chain fatty acid produced by strain SRB-530-5-HT was acetic acid. This strain tested positive for utilization of d-fructose, d-galacturonic acid, d-malic acid, l-alanyl l-threonine and l-glutamic acid but was negative for utilization of amygdalin, arbutin, α-d-glucose, 3-methyl d-glucose and salicin, in contrast to the type strain of the closest related species
Robinsoniella peoriensis
. The isolates were not able to use mannitol for growth. Based on genotypic, phenotypic and chemotaxonomic characteristics, we propose to create the new genus and species Murimonas intestini gen. nov., sp. nov. to accommodate the three strains SRB-530-5-HT ( = DSM 26524T = CCUG 63391T) (the type strain of Murimonas intestini), SRB-509-4-S-H ( = DSM 27577 = CCUG 64595) and SRB-524-4-S-H ( = DSM 27578 = CCUG 64594).
Collapse
|
130
|
Abstract
Celiac disease (CD) is a common chronic autoimmune enteropathy caused by gluten intake. To date, the only therapy for CD is the complete exclusion of dietary sources of grains and any food containing gluten. It has been hypothesized that the intestinal microbiota is somehow involved in CD. For this reason, probiotics are appearing as an interesting adjuvant in the dietetic management of CD. This review aims to discuss the characteristics of the microbiota in CD subjects and the use of probiotics as a novel therapy for CD. Comparisons between children with CD and controls show that their microbiota profiles differ; the former have fewer lactobacilli and bifidobacteria. Specific probiotics have been found to digest or alter gluten polypeptides. It has also been demonstrated that some bacterial species belonging to the genera Lactobacillus and Bifidobacterium exert protective properties on epithelial cells from damage caused by gliadin.
Collapse
|
131
|
No difference in small bowel microbiota between patients with irritable bowel syndrome and healthy controls. Sci Rep 2015; 5:8508. [PMID: 25687743 PMCID: PMC4330528 DOI: 10.1038/srep08508] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/22/2015] [Indexed: 12/17/2022] Open
Abstract
Several studies have indicated that colonic microbiota may exhibit important differences between patients with irritable bowel syndrome (IBS) and healthy controls. Less is known about the microbiota of the small bowel. We used massive parallel sequencing to explore the composition of small bowel mucosa-associated microbiota in patients with IBS and healthy controls. We analysed capsule biopsies from the jejunum of 35 patients (26 females) with IBS aged 18-(36)-57 years and 16 healthy volunteers (11 females) aged 20-(32)-48 years. Sequences were analysed based on taxonomic classification. The phyla with the highest total abundance across all samples were: Firmicutes (43%), Proteobacteria (23%), Bacteroidetes (15%), Actinobacteria (9.3%) and Fusobacteria (7.0%). The most abundant genera were: Streptococcus (19%), Veillonella (13%), Prevotella (12%), Rothia (6.4%), Haemophilus (5.7%), Actinobacillus (5.5%), Escherichia (4.6%) and Fusobacterium (4.3%). We found no difference among major phyla or genera between patients with IBS and controls. We identified a cluster of samples in the small bowel microbiota dominated by Prevotella, which may represent a common enterotype of the upper small intestine. The remaining samples formed a gradient, dominated by Streptococcus at one end and Escherichia at the other.
Collapse
|
132
|
Duda-Chodak A, Tarko T, Satora P, Sroka P. Interaction of dietary compounds, especially polyphenols, with the intestinal microbiota: a review. Eur J Nutr 2015; 54:325-41. [PMID: 25672526 PMCID: PMC4365176 DOI: 10.1007/s00394-015-0852-y] [Citation(s) in RCA: 363] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 01/30/2015] [Indexed: 02/08/2023]
Abstract
The intestinal microbiome plays an important role in the metabolism of chemical compounds found within food. Bacterial metabolites are different from those that can be generated by human enzymes because bacterial processes occur under anaerobic conditions and are based mainly on reactions of reduction and/or hydrolysis. In most cases, bacterial metabolism reduces the activity of dietary compounds; however, sometimes a specific product of bacterial transformation exhibits enhanced properties. Studies on the metabolism of polyphenols by the intestinal microbiota are crucial for understanding the role of these compounds and their impact on our health. This review article presents possible pathways of polyphenol metabolism by intestinal bacteria and describes the diet-derived bioactive metabolites produced by gut microbiota, with a particular emphasis on polyphenols and their potential impact on human health. Because the etiology of many diseases is largely correlated with the intestinal microbiome, a balance between the host immune system and the commensal gut microbiota is crucial for maintaining health. Diet-related and age-related changes in the human intestinal microbiome and their consequences are summarized in the paper.
Collapse
Affiliation(s)
- Aleksandra Duda-Chodak
- Department of Fermentation Technology and Technical Microbiology, Faculty of Food Technology, University of Agriculture in Krakow, ul. Balicka 122, 30-149, Kraków, Poland,
| | | | | | | |
Collapse
|
133
|
Duda-Chodak A, Tarko T, Satora P, Sroka P. Interaction of dietary compounds, especially polyphenols, with the intestinal microbiota: a review. Eur J Nutr 2015. [DOI: 10.1007/s00394-015-0852-y pmid: 25672526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
134
|
Gourbeyre P, Berri M, Lippi Y, Meurens F, Vincent-Naulleau S, Laffitte J, Rogel-Gaillard C, Pinton P, Oswald IP. Pattern recognition receptors in the gut: analysis of their expression along the intestinal tract and the crypt/villus axis. Physiol Rep 2015; 3:3/2/e12225. [PMID: 25677543 PMCID: PMC4393184 DOI: 10.14814/phy2.12225] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pattern recognition receptors (PRRs) play a critical role in the detection of microorganisms and the induction of inflammatory and immune responses. Using PCR and Western-blot analysis, this study investigated the differential expression in the intestine of 14 PRRs and nine associated cytokines. Thirty-two pigs were used to determine the expression of these markers (1) along the proximal/distal axis of the small intestine (duodenum, jejunum, and ileum) and (2) between the intestinal segments and their respective lymphoid organs (Peyer's patches [PP] and mesenteric lymph nodes [MLN]). Six additional animals were used to quantify the expression of these genes along the crypt/villus axis of jejunum, using microdissected samples. Most genes showed increased expression (1) in the distal than in the proximal parts of the small intestine (TLR3, 5, RIG-I, IL-1β, IL-8, and IFN-γ); (2) in lymphoid organs (TLR1, 2, 6, 9, 10, IL-10, TNF-α), especially the MLN (TLR4, 7, 8, NOD1, NOD2, NALP3, IFN-α, IL-6, IL-12, and TGF-β), than in intestinal segments. The analysis along the crypt/villus identified: (1) genes with higher expression in lamina propria (TLR1, 2, 4, 9, NOD1, NOD2, IL-1β, IL-10, TGF-β, TNF-α) and (2) genes with higher expression in the villus (TLR3, 5, 6, RIG-I, IL-6). These results highlight the differential expression of PRRs and cytokines along the proximal/distal and the crypt/villus axis of the intestine, contributing to a fine analysis of the complex functional architecture of the small intestine and should be related to the gut microbiota.
Collapse
Affiliation(s)
- Pascal Gourbeyre
- INRA, UMR1331, Toxalim, Toxicologie Alimentaire, Toulouse, France Université de Toulouse, INP, UMR1331, Toxalim, Toulouse, France
| | - Mustapha Berri
- INRA, UR1282, ISP, Infectiologie et Santé Publique, Nouzilly, France Université François Rabelais, UMR1282 Infectiologie et Santé Publique, Tours, France
| | - Yannick Lippi
- INRA, UMR1331, Toxalim, Toxicologie Alimentaire, Toulouse, France Université de Toulouse, INP, UMR1331, Toxalim, Toulouse, France
| | - François Meurens
- INRA, UR1282, ISP, Infectiologie et Santé Publique, Nouzilly, France Université François Rabelais, UMR1282 Infectiologie et Santé Publique, Tours, France Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Silvia Vincent-Naulleau
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, Jouy-en-Josas, France AgroParisTech, UMR1313 Génétique Animale et Biologie Intègrative, Jouy-en-Josas, France CEA, DSV, IRCM, Laboratoire de Radiobiologie et Etude du Génome, Domaine de Vilvert, Jouy-en-Josas, France
| | - Joëlle Laffitte
- INRA, UMR1331, Toxalim, Toxicologie Alimentaire, Toulouse, France Université de Toulouse, INP, UMR1331, Toxalim, Toulouse, France
| | - Claire Rogel-Gaillard
- INRA, UMR1313, Génétique Animale et Biologie Intégrative, Jouy-en-Josas, France AgroParisTech, UMR1313 Génétique Animale et Biologie Intègrative, Jouy-en-Josas, France
| | - Philippe Pinton
- INRA, UMR1331, Toxalim, Toxicologie Alimentaire, Toulouse, France Université de Toulouse, INP, UMR1331, Toxalim, Toulouse, France
| | - Isabelle P Oswald
- INRA, UMR1331, Toxalim, Toxicologie Alimentaire, Toulouse, France Université de Toulouse, INP, UMR1331, Toxalim, Toulouse, France
| |
Collapse
|
135
|
van den Bogert B, Meijerink M, Zoetendal EG, Wells JM, Kleerebezem M. Immunomodulatory properties of Streptococcus and Veillonella isolates from the human small intestine microbiota. PLoS One 2014; 9:e114277. [PMID: 25479553 PMCID: PMC4257559 DOI: 10.1371/journal.pone.0114277] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 11/09/2014] [Indexed: 12/14/2022] Open
Abstract
The human small intestine is a key site for interactions between the intestinal microbiota and the mucosal immune system. Here we investigated the immunomodulatory properties of representative species of commonly dominant small-intestinal microbial communities, including six streptococcal strains (four Streptococcus salivarius, one S. equinus, one S. parasanguinis) one Veillonella parvula strain, one Enterococcus gallinarum strain, and Lactobacillus plantarum WCFS1 as a bench mark strain on human monocyte-derived dendritic cells. The different streptococci induced varying levels of the cytokines IL-8, TNF-α, and IL-12p70, while the V. parvula strain showed a strong capacity to induce IL-6. E. gallinarum strain was a potent inducer of cytokines and TLR2/6 signalling. As Streptococcus and Veillonella can potentially interact metabolically and frequently co-occur in ecosystems, immunomodulation by pair-wise combinations of strains were also tested for their combined immunomodulatory properties. Strain combinations induced cytokine responses in dendritic cells that differed from what might be expected on the basis of the results obtained with the individual strains. A combination of (some) streptococci with Veillonella appeared to negate IL-12p70 production, while augmenting IL-8, IL-6, IL-10, and TNF-α responses. This suggests that immunomodulation data obtained in vitro with individual strains are unlikely to adequately represent immune responses to mixtures of gut microbiota communities in vivo. Nevertheless, analysing the immune responses of strains representing the dominant species in the intestine may help to identify immunomodulatory mechanisms that influence immune homeostasis.
Collapse
Affiliation(s)
- Bartholomeus van den Bogert
- Top Institute Food and Nutrition (TIFN), Wageningen, The Netherlands
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Marjolein Meijerink
- Host-Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands
| | - Erwin G. Zoetendal
- Top Institute Food and Nutrition (TIFN), Wageningen, The Netherlands
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Jerry M. Wells
- Host-Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands
| | - Michiel Kleerebezem
- Top Institute Food and Nutrition (TIFN), Wageningen, The Netherlands
- Host-Microbe Interactomics Group, Wageningen University, Wageningen, The Netherlands
- NIZO Food Research B.V., Ede, The Netherlands
- * E-mail:
| |
Collapse
|
136
|
Wacklin P, Laurikka P, Lindfors K, Collin P, Salmi T, Lähdeaho ML, Saavalainen P, Mäki M, Mättö J, Kurppa K, Kaukinen K. Altered duodenal microbiota composition in celiac disease patients suffering from persistent symptoms on a long-term gluten-free diet. Am J Gastroenterol 2014; 109:1933-41. [PMID: 25403367 DOI: 10.1038/ajg.2014.355] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/02/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVES A significant fraction of celiac disease patients suffer from persistent symptoms despite a long-term gluten-free diet (GFD) and normalized small bowel mucosa. The commonly suggested reasons, such as inadvertent gluten-intake or presence of other gastrointestinal disease, do not explain the symptoms in all these patients. Recently, alterations in intestinal microbiota have been associated with autoimmune disorders, including celiac disease. This led us to test a hypothesis that abnormal intestinal microbiota may be associated with persisting gastrointestinal symptoms in treated celiac disease patients. METHODS Duodenal microbiota was analyzed in 18 GFD-treated patients suffering from persistent symptoms and 18 treated patients without symptoms by 16S rRNA gene pyrosequencing. The celiac disease patients had been following a strict GFD for several years and had restored small bowel mucosa and negative celiac autoantibodies. Their symptoms on GFD were assessed with Gastrointestinal Symptom Rating Scale. RESULTS The results of several clustering methods showed that the treated celiac disease patients with persistent symptoms were colonized by different duodenal microbiota in comparison with patients without symptoms. The treated patients with persistent symptoms had a higher relative abundance of Proteobacteria (P=0.04) and a lower abundance of Bacteroidetes (P=0.01) and Firmicutes (P=0.05). Moreover, their microbial richness was reduced. The results indicated intestinal dysbiosis in patients with persistent symptoms even while adhering to a strict GFD. CONCLUSIONS Our findings indicate that dysbiosis of microbiota is associated with persistent gastrointestinal symptoms in treated celiac disease patients and open new possibilities to treat this subgroup of patients.
Collapse
Affiliation(s)
| | - Pilvi Laurikka
- School of Medicine, University of Tampere, Tampere, Finland
| | - Katri Lindfors
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Pekka Collin
- 1] School of Medicine, University of Tampere, Tampere, Finland [2] Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Teea Salmi
- 1] School of Medicine, University of Tampere, Tampere, Finland [2] Department of Dermatology, Tampere University Hospital, Tampere, Finland
| | - Marja-Leena Lähdeaho
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Päivi Saavalainen
- Research Programs Unit, Immunobiology, and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Markku Mäki
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Jaana Mättö
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - Kalle Kurppa
- Tampere Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Katri Kaukinen
- 1] School of Medicine, University of Tampere, Tampere, Finland [2] Department of Internal Medicine, Tampere University Hospital, Tampere and Seinäjoki Central Hospital, Seinäjoki, Finland
| |
Collapse
|
137
|
Pediatric small intestine bacterial overgrowth in low-income countries. Trends Mol Med 2014; 21:6-15. [PMID: 25486880 DOI: 10.1016/j.molmed.2014.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/07/2014] [Accepted: 11/11/2014] [Indexed: 02/08/2023]
Abstract
Small intestine bacterial overgrowth (SIBO) occurs when colonic quantities of commensal bacteria are present in the small bowel. SIBO is associated with conditions of disrupted gastrointestinal (GI) motility leading to stasis of luminal contents. Recent data show that SIBO is also found in children living in unsanitary conditions who do not have access to clean water. SIBO leads to impaired micronutrient absorption and increased GI permeability, both of which may contribute to growth stunting in children. SIBO also disrupts mucosal immunity and has been implicated in oral vaccination underperformance and the development of celiac disease. SIBO in the setting of the impoverished human habitats may be an under-recognized cause of pediatric morbidity and mortality in the developing world.
Collapse
|
138
|
Delorme C, Abraham AL, Renault P, Guédon E. Genomics of Streptococcus salivarius, a major human commensal. INFECTION GENETICS AND EVOLUTION 2014; 33:381-92. [PMID: 25311532 DOI: 10.1016/j.meegid.2014.10.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/30/2014] [Accepted: 10/02/2014] [Indexed: 10/24/2022]
Abstract
The salivarius group of streptococci is of particular importance for humans. This group consists of three genetically similar species, Streptococcus salivarius, Streptococcus vestibularis and Streptococcus thermophilus. S. salivarius and S. vestibularis are commensal organisms that may occasionally cause opportunistic infections in humans, whereas S. thermophilus is a food bacterium widely used in dairy production. We developed Multilocus sequence typing (MLST) and comparative genomic analysis to confirm the clear separation of these three species. These analyses also identified a subgroup of four strains, with a core genome diverging by about 10%, in terms of its nucleotide sequence, from that of S. salivarius sensu stricto. S. thermophilus species displays a low level of nucleotide variability, due to its recent emergence with the development of agriculture. By contrast, nucleotide variability is high in the other two species of the salivarius group, reflecting their long-standing association with humans. The species of the salivarius group have genome sizes ranging from the smallest (∼ 1.7 Mb for S. thermophilus) to the largest (∼ 2.3 Mb for S. salivarius) among streptococci, reflecting genome reduction linked to a narrow, nutritionally rich environment for S. thermophilus, and natural, more competitive niches for the other two species. Analyses of genomic content have indicated that the core genes of S. salivarius account for about two thirds of the genome, indicating considerable variability of gene content and differences in potential adaptive features. Furthermore, we showed that the genome of this species is exceptionally rich in genes encoding surface factors, glycosyltransferases and response regulators. Evidence of widespread genetic exchanges was obtained, probably involving a natural competence system and the presence of diverse mobile elements. However, although the S. salivarius strains studied were isolated from several human body-related sites (all levels of the digestive tract, skin, breast milk, and body fluids) and included clinical strains, no genetic or genomic niche-specific features could be identified to discriminate specific group.
Collapse
Affiliation(s)
- Christine Delorme
- INRA, UMR 1319 Micalis, Domaine de Vilvert, F-78352 Jouy-en-Josas, France; AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Anne-Laure Abraham
- INRA, UMR 1319 Micalis, Domaine de Vilvert, F-78352 Jouy-en-Josas, France; AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Pierre Renault
- INRA, UMR 1319 Micalis, Domaine de Vilvert, F-78352 Jouy-en-Josas, France; AgroParisTech, UMR MICALIS, Jouy-en-Josas, France
| | - Eric Guédon
- INRA, UMR 1319 Micalis, Domaine de Vilvert, F-78352 Jouy-en-Josas, France; AgroParisTech, UMR MICALIS, Jouy-en-Josas, France.
| |
Collapse
|
139
|
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: 745] [Impact Index Per Article: 67.7] [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
|
140
|
Kim JM. [Roles of enteric microbial composition and metabolism in health and diseases]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2014; 62:191-205. [PMID: 24162706 DOI: 10.4166/kjg.2013.62.4.191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A complex microbiota colonizes mucosal layers in different regions of the human gut. In the healthy state, the microbial communities provide nutrients and energy to the host via fermentation of non-digestible dietary components in the large intestine. In contrast, they can play roles in inflammation and infection, including gastrointestinal diseases and metabolic syndrome such as obesity. However, because of the complexity of the microbial community, the functional connections between the enteric microbiota and metabolism are less well understood. Nevertheless, major progress has been made in defining dominant bacterial species, community profiles, and systemic characteristics that produce stable microbiota beneficial to health, and in identifying their roles in enteric metabolism. Through studies in both mice and humans, we are recently in a better position to understand what effect the enteric microbiota has on the metabolism by improving energy yield from food and modulating dietary components. Achieving better knowledge of this information may provide insights into new possibilities that reconstitution of enteric microbiota via diet can provide the maintenance of healthy state and therapeutic/preventive strategies against metabolic syndrome such as obesity. This review focuses on enteric microbial composition and metabolism on healthy and diseased states.
Collapse
Affiliation(s)
- Jung Mogg Kim
- Department of Microbiology, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
141
|
Role of the lower and upper intestine in the production and absorption of gut microbiota-derived PUFA metabolites. PLoS One 2014; 9:e87560. [PMID: 24475308 PMCID: PMC3903770 DOI: 10.1371/journal.pone.0087560] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/21/2013] [Indexed: 01/01/2023] Open
Abstract
In vitro studies have suggested that isolated gut bacteria are able to metabolize PUFA into CLA (conjugated linoleic acids) and CLnA (conjugated linolenic acids). However, the bioavailability of fatty acid metabolites produced in vivo by the gut microbes remains to be studied. Therefore, we measured intestinal concentration and plasma accumulation of bacterial metabolites produced from dietary PUFA in mice, first injected with a lipoprotein lipase inhibitor, then force-fed with either sunflower oil (200 µl) rich in n-6 PUFA or linseed oil (200 µl) rich in n-3 PUFA. The greatest production of bacterial metabolites was observed in the caecum and colon, and at a much lesser extent in the jejunum and ileum. In the caecal content, CLA proportions were higher in sunflower oil force-fed mice whereas CLnA proportions were higher in linseed oil force-fed mice. The accumulation of the main metabolites (CLA cis-9,trans-11-18:2 and CLnA cis-9,trans-11,cis-15-18:3) in the caecal tissue was not associated with their increase in the plasma, therefore suggesting that, if endogenously produced CLA and CLnA have any biological role in host metabolism regulation, their effect would be confined at the intestinal level, where the microbiota is abundant.
Collapse
|
142
|
Kohl KD, Miller AW, Marvin JE, Mackie R, Dearing MD. Herbivorous rodents (Neotoma spp.) harbour abundant and active foregut microbiota. Environ Microbiol 2014; 16:2869-78. [DOI: 10.1111/1462-2920.12376] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/13/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Kevin D. Kohl
- Department of Biology; University of Utah; Salt Lake City UT 84112 USA
| | - Aaron W. Miller
- Department of Biology; University of Utah; Salt Lake City UT 84112 USA
| | - James E. Marvin
- Flow Cytometry Core Facility; University of Utah; Salt Lake City UT 84132 USA
| | - Roderick Mackie
- Department of Animal Sciences; University of Illinois; Urbana IL 61801 USA
| | - M. Denise Dearing
- Department of Biology; University of Utah; Salt Lake City UT 84112 USA
| |
Collapse
|
143
|
Håkansson Å, Tormo-Badia N, Baridi A, Xu J, Molin G, Hagslätt ML, Karlsson C, Jeppsson B, Cilio CM, Ahrné S. Immunological alteration and changes of gut microbiota after dextran sulfate sodium (DSS) administration in mice. Clin Exp Med 2014; 15:107-20. [PMID: 24414342 PMCID: PMC4308640 DOI: 10.1007/s10238-013-0270-5] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/14/2013] [Indexed: 12/17/2022]
Abstract
Ulcerative colitis (UC) is characterized by chronic inflammation of the colonic mucosa. Administration of dextran sulfate sodium (DSS) to animals is a frequently used model to mimic human colitis. Deregulation of the immune response to the enteric microflora or pathogens as well as increased intestinal permeability have been proposed as disease-driving mechanisms. To enlarge the understanding of the pathogenesis, we have studied the effect of DSS on the immune system and gut microbiota in mice. Intestinal inflammation was verified through histological evaluation and myeloperoxidase activity. Immunological changes were assessed by flow cytometry in spleen, Peyer′s patches and mesenteric lymph nodes and through multiplex cytokine profiling. In addition, quantification of the total amount of bacteria on colonic mucosa as well as the total amount of lactobacilli, Akkermansia, Desulfovibrio and Enterobacteriaceae was performed by the use of quantitative PCR. Diversity and community structure were analysed by terminal restriction fragment length polymorphism (T-RFLP) patterns, and principal component analysis was utilized on immunological and T-RFLP patterns. DSS-induced colitis show clinical and histological similarities to UC. The composition of the colonic microflora was profoundly changed and correlated with several alterations of the immune system. The results demonstrate a relationship between multiple immunological changes and alterations of the gut microbiota after DSS administration. These data highlight and improve the definition of the immunological basis of the disease and suggest a role for dysregulation of the gut microbiota in the pathogenesis of colitis.
Collapse
Affiliation(s)
- Å Håkansson
- Food Hygiene, Division of Applied Nutrition and Food Chemistry, Lund University, Lund, Sweden,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Turroni F, Ventura M, Buttó LF, Duranti S, O’Toole PW, Motherway MO, van Sinderen D. Molecular dialogue between the human gut microbiota and the host: a Lactobacillus and Bifidobacterium perspective. Cell Mol Life Sci 2014; 71:183-203. [PMID: 23516017 PMCID: PMC11113728 DOI: 10.1007/s00018-013-1318-0] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 02/13/2013] [Accepted: 03/04/2013] [Indexed: 02/06/2023]
Abstract
The human gut represents a highly complex ecosystem, which is densely colonized by a myriad of microorganisms that influence the physiology, immune function and health status of the host. Among the many members of the human gut microbiota, there are microorganisms that have co-evolved with their host and that are believed to exert health-promoting or probiotic effects. Probiotic bacteria isolated from the gut and other environments are commercially exploited, and although there is a growing list of health benefits provided by the consumption of such probiotics, their precise mechanisms of action have essentially remained elusive. Genomics approaches have provided exciting new opportunities for the identification of probiotic effector molecules that elicit specific responses to influence the physiology and immune function of their human host. In this review, we describe the current understanding of the intriguing relationships that exist between the human gut and key members of the gut microbiota such as bifidobacteria and lactobacilli, discussed here as prototypical groups of probiotic microorganisms.
Collapse
Affiliation(s)
- Francesca Turroni
- Alimentary Pharmabiotic Centre, Department of Microbiology Biosciences Institute, University College Cork, National University of Ireland, Western Road, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Genetics, Biology of Microorganisms, Anthropology and Evolution, University of Parma, Parma, Italy
| | - Ludovica F. Buttó
- Alimentary Pharmabiotic Centre, Department of Microbiology Biosciences Institute, University College Cork, National University of Ireland, Western Road, Cork, Ireland
| | - Sabrina Duranti
- Laboratory of Probiogenomics, Department of Genetics, Biology of Microorganisms, Anthropology and Evolution, University of Parma, Parma, Italy
| | - Paul W. O’Toole
- Alimentary Pharmabiotic Centre, Department of Microbiology Biosciences Institute, University College Cork, National University of Ireland, Western Road, Cork, Ireland
| | - Mary O’Connell Motherway
- Alimentary Pharmabiotic Centre, Department of Microbiology Biosciences Institute, University College Cork, National University of Ireland, Western Road, Cork, Ireland
| | - Douwe van Sinderen
- Alimentary Pharmabiotic Centre, Department of Microbiology Biosciences Institute, University College Cork, National University of Ireland, Western Road, Cork, Ireland
| |
Collapse
|
145
|
Draft Genome Sequence of Enterococcus sp. Strain HSIEG1, Isolated from the Human Small Intestine. GENOME ANNOUNCEMENTS 2013; 1:1/6/e01013-13. [PMID: 24336366 PMCID: PMC3861419 DOI: 10.1128/genomea.01013-13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Enterococcus sp. strain HSIEG1 was isolated from the human small intestine. Its draft genome predicts a broad carbohydrate fermentation capability, which matches well with the observed physiological characteristics of this strain. This metabolic flexibility is expected to be of importance for survival and growth in the small intestinal habitat.
Collapse
|
146
|
Moossavi S, Zhang H, Sun J, Rezaei N. Host-microbiota interaction and intestinal stem cells in chronic inflammation and colorectal cancer. Expert Rev Clin Immunol 2013; 9:409-22. [PMID: 23634736 DOI: 10.1586/eci.13.27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Inflammatory bowel disease (IBD) and colorectal cancer (CRC) are the major diseases of the lower gastrointestinal tract. The intestinal epithelium plays a critical role in the host's interactions with the large communities of resident luminal bacteria. Epithelial cells recognize the bacterial components via pattern-recognition receptors. Toll-like receptors (TLRs) are a major class of pattern-recognition receptors that are present on intestinal epithelial cells, including putative stem cells. Stem cells are responsible for tissue homeostasis and regeneration after injury including IBD. Stem cells are also implicated in the pathogenesis of CRC. In susceptible individuals, disruption of normal homeostatic balance between the host's mucosal cells and enteric microflora is believed to result in aberrant immune responses against the resident commensal bacteria, leading to IBD. Microbiological analyses have revealed that the composition and localization of microbiota is altered in CRC and IBD. It is plausible that stem cells directly sense and respond to microbiota. This review aims to summarize the current knowledge on the effect of microbiota and TLR signaling on intestinal stem cells. It also describes how TLR signaling could affect the stem cell regulatory pathways.
Collapse
Affiliation(s)
- Shirin Moossavi
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
147
|
Anti-inflammatory properties of Streptococcus salivarius, a commensal bacterium of the oral cavity and digestive tract. Appl Environ Microbiol 2013; 80:928-34. [PMID: 24271166 DOI: 10.1128/aem.03133-13] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Streptococcus salivarius is one of the first colonizers of the human oral cavity and gut after birth and therefore may contribute to the establishment of immune homeostasis and regulation of host inflammatory responses. The anti-inflammatory potential of S. salivarius was first evaluated in vitro on human intestinal epithelial cells and human peripheral blood mononuclear cells. We show that live S. salivarius strains inhibited in vitro the activation of the NF-κB pathway on intestinal epithelial cells. We also demonstrate that the live S. salivarius JIM8772 strain significantly inhibited inflammation in severe and moderate colitis mouse models. These in vitro and in vivo anti-inflammatory properties were not found with heat-killed S. salivarius, suggesting a protective response exclusively with metabolically active bacteria.
Collapse
|
148
|
Lavelle A, Lennon G, Docherty N, Balfe A, Mulcahy HE, Doherty G, O′Donoghue D, Hyland JM, Shanahan F, Sheahan K, Coffey JC, Winter DC, O′Connell PR. Depth-dependent differences in community structure of the human colonic microbiota in health. PLoS One 2013; 8:e78835. [PMID: 24223167 PMCID: PMC3819420 DOI: 10.1371/journal.pone.0078835] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 09/16/2013] [Indexed: 12/21/2022] Open
Abstract
Objective The aims of this study were to develop techniques for spatial microbial assessment in humans and to establish colonic luminal and mucosal spatial ecology, encompassing longitudinal and cross-sectional axes. Design A microbiological protected specimen brush was used in conjunction with a biopsy forceps to sample the colon in nine healthy volunteers undergoing colonoscopy. Terminal Restriction Fragment Length Polymorphism analysis was used to determine the major variables in the spatial organization of the colonic microbiota. Results Protected Specimen Brush sampling retrieved region-specific, uncontaminated samples that were enriched for bacterial DNA and depleted in human DNA when compared to biopsy samples. Terminal Restriction Fragment Length Polymorphism analysis revealed a segmentation of bacterial communities between the luminal brush and biopsy-associated ecological niches with little variability across the longitudinal axis of the colon and reduced diversity in brush samples. Conclusion These results support the concept of a microbiota with little longitudinal variability but with some degree of segregation between luminal and mucosal communities.
Collapse
Affiliation(s)
- Aonghus Lavelle
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Grainne Lennon
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Neil Docherty
- Department of Physiology, Trinity College Dublin, Ireland
| | - Aine Balfe
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
| | - Hugh E. Mulcahy
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Glen Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Diarmuid O′Donoghue
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - John M. Hyland
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Fergus Shanahan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Kieran Sheahan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - J. Calvin Coffey
- 4i Centre for Interventions in Inflammation, Infection and Immunity, Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - Desmond C. Winter
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - P. Ronan O′Connell
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- * E-mail:
| |
Collapse
|
149
|
Zhang Z, Geng J, Tang X, Fan H, Xu J, Wen X, Ma ZS, Shi P. Spatial heterogeneity and co-occurrence patterns of human mucosal-associated intestinal microbiota. ISME JOURNAL 2013; 8:881-93. [PMID: 24132077 DOI: 10.1038/ismej.2013.185] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 09/06/2013] [Accepted: 09/17/2013] [Indexed: 01/19/2023]
Abstract
Human gut microbiota shows high inter-subject variations, but the actual spatial distribution and co-occurrence patterns of gut mucosa microbiota that occur within a healthy human instestinal tract remain poorly understood. In this study, we illustrated a model of this mucosa bacterial communities' biogeography, based on the largest data set so far, obtained via 454-pyrosequencing of bacterial 16S rDNAs associated with 77 matched biopsy tissue samples taken from terminal ileum, ileocecal valve, ascending colon, transverse colon, descending colon, sigmoid colon and rectum of 11 healthy adult subjects. Borrowing from macro-ecology, we used both Taylor's power law analysis and phylogeny-based beta-diversity metrics to uncover a highly heterogeneous distribution pattern of mucosa microbial inhabitants along the length of the intestinal tract. We then developed a spatial dispersion model with an R-squared value greater than 0.950 to map out the gut mucosa-associated flora's non-linear spatial distribution pattern for 51.60% of the 188 most abundant gut bacterial species. Furthermore, spatial co-occurring network analysis of mucosa microbial inhabitants together with occupancy (that is habitat generalists, specialists and opportunist) analyses implies that ecological relationships (both oppositional and symbiotic) between mucosa microbial inhabitants may be important contributors to the observed spatial heterogeneity of mucosa microbiota along the human intestine and may even potentially be associated with mutual cooperation within and functional stability of the gut ecosystem.
Collapse
Affiliation(s)
- Zhigang Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jiawei Geng
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Xiaodan Tang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Hong Fan
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Jinchao Xu
- Center for Computational Mathematics and Applications, Department of Mathematics, Pennsylvania State University, University Park, PA, USA
| | - Xiujun Wen
- College of Forestry, South China Agricultural University, Guangzhou, China
| | - Zhanshan Sam Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Peng Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
150
|
Xue LY, Ouyang Q, Zhou XG, Huang ZH, Chen W, Chen M, Yu LM. Bacterial immune interaction in experimental colitis. J Dig Dis 2013; 14:526-35. [PMID: 23734583 DOI: 10.1111/1751-2980.12079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES This study aimed to analyze the effects of 5-aminosalicylic acid (5-ASA) on intestinal microbiota and immune regulation in inflammatory bowel disease (IBD) and to investigate the correlation between intestinal microbiota and immune factors. METHODS Colitis in mice was induced by oxazolone. The community composition of luminal and mucosal microbiota was analyzed by a terminal restriction fragment length polymorphism. The expression of occludin, toll-like receptor (TLR)-2, TLR-4 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 proteins were measured by immunohistochemistry and Western blot. Linear correlation between intestinal microbial community and the severity of the colitis or intestinal microbial community and expressions of immune factors were determined. RESULTS Protective bacteria decreased while aggressive bacteria increased in the colitis group. The richness and diversity of both luminal and mucosal microbiota decreased in the colitis group the decrease was enhanced in the 5-ASA-treated group. The diversity of mucosal microbiota significantly correlated with the extent of the colitis. Expressions of occludin, TLR-2, TLR-4, tumor necrosis factor-α and NF-κB p65 were significantly correlated with the diversity of mucosal microbiota. CONCLUSIONS Mucosal microbiota are important in the pathogenesis of IBD. 5-ASA increases protective bacteria but decreases aggressive bacteria, thus inducing the new intestinal microbial homeostasis.
Collapse
Affiliation(s)
- Lin Yun Xue
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | | | | | | | | | | | | |
Collapse
|