1251
|
Wu XM, Tan RX. Interaction between gut microbiota and ethnomedicine constituents. Nat Prod Rep 2019; 36:788-809. [DOI: 10.1039/c8np00041g] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This highlight reviews the interaction processes between gut microbiota and ethnomedicine constituents, which may conceptualize future therapeutic strategies.
Collapse
Affiliation(s)
- Xue Ming Wu
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Ren Xiang Tan
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
- State Key Laboratory of Pharmaceutical Biotechnology
| |
Collapse
|
1252
|
Harrison CA, Laubitz D, Midura-Kiela MT, Jamwal DR, Besselsen DG, Ghishan FK, Kiela PR. Sexual Dimorphism in the Response to Broad-spectrum Antibiotics During T Cell-mediated Colitis. J Crohns Colitis 2019; 13:115-126. [PMID: 30252029 PMCID: PMC6302957 DOI: 10.1093/ecco-jcc/jjy144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Broad-spectrum antibiotics [Abx], including combination therapy with ciprofloxacin and metronidazole, are often prescribed during the treatment of inflammatory bowel disease [IBD] to alleviate symptoms, but with varying success. In this pilot study, we studied the effects of Abx on the course of experimental colitis, with a particular focus on sex as a determinant of the microbial and inflammatory responses. METHODS The effects of Abx were tested on colonic inflammation and microbiome in male and female Rag-/- mice, using adoptive transfer of naïve T cells to induce colitis in a short-term [2-week] and long-term [9-week] study. RESULTS We observed disparities between the sexes in both the response to adoptive T cell transfer and the effects of Abx. At baseline without Abx, female mice displayed a trend toward a more severe colitis than males. In both the short- and the long-term experiments, gut microbiota of some female mice exposed to Abx showed weak, delayed, or negligible shifts. Caecum weight was significantly lower in Abx-treated females. Abx exposure favoured a quick and persistent rise in Enterococcaceae exclusively in females. Males had higher relative abundance of Lactobacillaceae following Abx exposure relative to females. Abx-treated females trended toward higher colitis scores than Abx-treated males, and towards higher levels of IL-17A, NOS2, and IL-22. CONCLUSIONS Although preliminary, our results suggest a differential response to both inflammation and Abx between male and female mice, The findings may be relevant to current practice and also as the basis for further studies on the differential gender effects during long-term antibiotic exposure in IBD.
Collapse
Affiliation(s)
- Christy A Harrison
- Department of Pediatrics, Steele Children’s Research Center, Tucson, AZ, USA
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Daniel Laubitz
- Department of Pediatrics, Steele Children’s Research Center, Tucson, AZ, USA
| | | | - Deepa R Jamwal
- Department of Pediatrics, Steele Children’s Research Center, Tucson, AZ, USA
| | | | - Fayez K Ghishan
- Department of Pediatrics, Steele Children’s Research Center, Tucson, AZ, USA
| | - Pawel R Kiela
- Department of Pediatrics, Steele Children’s Research Center, Tucson, AZ, USA
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
1253
|
Tyakht AV, Manolov AI, Kanygina AV, Ischenko DS, Kovarsky BA, Popenko AS, Pavlenko AV, Elizarova AV, Rakitina DV, Baikova JP, Ladygina VG, Kostryukova ES, Karpova IY, Semashko TA, Larin AK, Grigoryeva TV, Sinyagina MN, Malanin SY, Shcherbakov PL, Kharitonova AY, Khalif IL, Shapina MV, Maev IV, Andreev DN, Belousova EA, Buzunova YM, Alexeev DG, Govorun VM. Genetic diversity of Escherichia coli in gut microbiota of patients with Crohn's disease discovered using metagenomic and genomic analyses. BMC Genomics 2018; 19:968. [PMID: 30587114 PMCID: PMC6307143 DOI: 10.1186/s12864-018-5306-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Crohn's disease is associated with gut dysbiosis. Independent studies have shown an increase in the abundance of certain bacterial species, particularly Escherichia coli with the adherent-invasive pathotype, in the gut. The role of these species in this disease needs to be elucidated. METHODS We performed a metagenomic study investigating the gut microbiota of patients with Crohn's disease. A metagenomic reconstruction of the consensus genome content of the species was used to assess the genetic variability. RESULTS The abnormal shifts in the microbial community structures in Crohn's disease were heterogeneous among the patients. The metagenomic data suggested the existence of multiple E. coli strains within individual patients. We discovered that the genetic diversity of the species was high and that only a few samples manifested similarity to the adherent-invasive varieties. The other species demonstrated genetic diversity comparable to that observed in the healthy subjects. Our results were supported by a comparison of the sequenced genomes of isolates from the same microbiota samples and a meta-analysis of published gut metagenomes. CONCLUSIONS The genomic diversity of Crohn's disease-associated E. coli within and among the patients paves the way towards an understanding of the microbial mechanisms underlying the onset and progression of the Crohn's disease and the development of new strategies for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Alexander V. Tyakht
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
- ITMO University, 49 Kronverkskiy pr, Saint-Petersburg, Russian Federation 197101
| | - Alexander I. Manolov
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Alexandra V. Kanygina
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
| | - Dmitry S. Ischenko
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
| | - Boris A. Kovarsky
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Anna S. Popenko
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Alexander V. Pavlenko
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Anna V. Elizarova
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
| | - Daria V. Rakitina
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Julia P. Baikova
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Valentina G. Ladygina
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Elena S. Kostryukova
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
| | - Irina Y. Karpova
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Tatyana A. Semashko
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
| | - Andrei K. Larin
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | | | - Mariya N. Sinyagina
- Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008
| | - Sergei Y. Malanin
- Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008
| | - Petr L. Shcherbakov
- Moscow Clinical Scientific Center, 86 Shosse Entuziastov St., Moscow, Russian Federation 111123
| | - Anastasiya Y. Kharitonova
- Clinical and Research Institute of Emergency Children’s Surgery and Trauma, 22 Bolshaya Polyanka St., Moscow, Russian Federation 119180
| | - Igor L. Khalif
- State Scientific Center of Coloproctology, 2 Salam Adil St., Moscow, Russian Federation 123423
| | - Marina V. Shapina
- State Scientific Center of Coloproctology, 2 Salam Adil St., Moscow, Russian Federation 123423
| | - Igor V. Maev
- Moscow State University of Medicine and Dentistry, Build. 6, 20 Delegatskaya St., Moscow, Russian Federation 127473
| | - Dmitriy N. Andreev
- Moscow State University of Medicine and Dentistry, Build. 6, 20 Delegatskaya St., Moscow, Russian Federation 127473
| | - Elena A. Belousova
- Moscow Regional Research and Clinical Institute, 61/2 Shchepkina str, Moscow, Russian Federation 129110
| | - Yulia M. Buzunova
- Moscow Regional Research and Clinical Institute, 61/2 Shchepkina str, Moscow, Russian Federation 129110
| | - Dmitry G. Alexeev
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
| | - Vadim M. Govorun
- Federal Research and Clinical Centre of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy per., Dolgoprudny, Moscow Region, Russian Federation 141700
- M.M. Shemyakin - Yu.A. Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow, Russian Federation 117997
| |
Collapse
|
1254
|
Fu X, Liu Z, Zhu C, Mou H, Kong Q. Nondigestible carbohydrates, butyrate, and butyrate-producing bacteria. Crit Rev Food Sci Nutr 2018; 59:S130-S152. [PMID: 30580556 DOI: 10.1080/10408398.2018.1542587] [Citation(s) in RCA: 317] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nondigestible carbohydrates (NDCs) are fermentation substrates in the colon after escaping digestion in the upper gastrointestinal tract. Among NDCs, resistant starch is not hydrolyzed by pancreatic amylases but can be degraded by enzymes produced by large intestinal bacteria, including clostridia, bacteroides, and bifidobacteria. Nonstarch polysaccharides, such as pectin, guar gum, alginate, arabinoxylan, and inulin fructans, and nondigestible oligosaccharides and their derivatives, can also be fermented by beneficial bacteria in the large intestine. Butyrate is one of the most important metabolites produced through gastrointestinal microbial fermentation and functions as a major energy source for colonocytes by directly affecting the growth and differentiation of colonocytes. Moreover, butyrate has various physiological effects, including enhancement of intestinal barrier function and mucosal immunity. In this review, several representative NDCs are introduced, and their chemical components, structures, and physiological functions, including promotion of the proliferation of butyrate-producing bacteria and enhancement of butyrate production, are discussed. We also describe the strategies for achieving directional accumulation of colonic butyrate based on endogenous generation mechanisms.
Collapse
Affiliation(s)
- Xiaodan Fu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Zhemin Liu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Changliang Zhu
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Haijin Mou
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| | - Qing Kong
- a College of Food Science and Engineering , Ocean University of China , Qingdao , China
| |
Collapse
|
1255
|
The gut microbiome is required for full protection against acute arsenic toxicity in mouse models. Nat Commun 2018; 9:5424. [PMID: 30575732 PMCID: PMC6303300 DOI: 10.1038/s41467-018-07803-9] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 11/21/2018] [Indexed: 01/09/2023] Open
Abstract
Arsenic poisons an estimated 200 million people worldwide through contaminated food and drinking water. Confusingly, the gut microbiome has been suggested to both mitigate and exacerbate arsenic toxicity. Here, we show that the microbiome protects mice from arsenic-induced mortality. Both antibiotic-treated and germ-free mice excrete less arsenic in stool and accumulate more arsenic in organs compared to control mice. Mice lacking the primary arsenic detoxification enzyme (As3mt) are hypersensitive to arsenic after antibiotic treatment or when derived germ-free, compared to wild-type and/or conventional counterparts. Human microbiome (stool) transplants protect germ-free As3mt-KO mice from arsenic-induced mortality, but protection depends on microbiome stability and the presence of specific bacteria, including Faecalibacterium. Our results demonstrate that both a functional As3mt and specific microbiome members are required for protection against acute arsenic toxicity in mouse models. We anticipate that the gut microbiome will become an important explanatory factor of disease (arsenicosis) penetrance in humans, and a novel target for prevention and treatment strategies. It is unclear whether the gut microbiome can mitigate or exacerbate arsenic toxicity. Here, Coryell et al. show that the human gut microbiome protects mice from arsenic-induced mortality, with protection levels correlating with the relative abundance of the human commensal Faecalibacterium.
Collapse
|
1256
|
Barone F, Laghi L, Gianotti A, Ventrella D, Saa DLT, Bordoni A, Forni M, Brigidi P, Bacci ML, Turroni S. In Vivo Effects of Einkorn Wheat (Triticum monococcum) Bread on the Intestinal Microbiota, Metabolome, and on the Glycemic and Insulinemic Response in the Pig Model. Nutrients 2018; 11:16. [PMID: 30577558 PMCID: PMC6356388 DOI: 10.3390/nu11010016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 01/08/2023] Open
Abstract
Einkorn wheat (Triticum monococcum) is characterized by high content of proteins, bioactive compounds, such as polyunsaturated fatty acids, fructans, tocols, carotenoids, alkylresorcinols, and phytosterols, and lower α-, β-amylase and lipoxygenase activities compared to polyploid wheat. These features make einkorn flour a good candidate to provide healthier foods. In the present study, we investigated the effects of einkorn bread (EB) on the intestinal physiology and metabolism of the pig model by characterizing the glycemic and insulinemic response, and the microbiota and metabolome profiles. Sixteen commercial hybrid pigs were enrolled in the study; four pigs were used to characterize postprandial glycemic and insulinemic responses and twelve pigs underwent a 30-day dietary intervention to assess microbiota and metabolome changes after EB or standard wheat bread (WB) consumption. The postprandial insulin rise after an EB meal was characterized by a lower absolute level, and, as also observed for glucose, by a biphasic shape in contrast to that in response to a WB meal. The consumption of EB led to enrichment in short-chain fatty acid producers (e.g., Blautia, Faecalibacterium, and Oscillospira) in the gut microbiota and to higher metabolic diversity with lower content of succinate, probably related to improved absorption and therefore promoting intestinal gluconeogenesis. The observed changes, at both a compositional and metabolic scale, strongly suggest that EB consumption may support a health-promoting configuration of the intestinal ecosystem.
Collapse
Affiliation(s)
- Francesca Barone
- Department of Veterinary Medical Science, University of Bologna, 40064 Ozzano dell'Emilia, Italy.
| | - Luca Laghi
- Department of Agro-Food Science and Technology, University of Bologna, 47521 Cesena, Italy.
| | - Andrea Gianotti
- Department of Agro-Food Science and Technology, University of Bologna, 47521 Cesena, Italy.
| | - Domenico Ventrella
- Department of Veterinary Medical Science, University of Bologna, 40064 Ozzano dell'Emilia, Italy.
| | | | - Alessandra Bordoni
- Department of Agro-Food Science and Technology, University of Bologna, 47521 Cesena, Italy.
| | - Monica Forni
- Department of Veterinary Medical Science, University of Bologna, 40064 Ozzano dell'Emilia, Italy.
| | - Patrizia Brigidi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy.
| | - Maria Laura Bacci
- Department of Veterinary Medical Science, University of Bologna, 40064 Ozzano dell'Emilia, Italy.
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
1257
|
Fitzgerald CB, Shkoporov AN, Sutton TDS, Chaplin AV, Velayudhan V, Ross RP, Hill C. Comparative analysis of Faecalibacterium prausnitzii genomes shows a high level of genome plasticity and warrants separation into new species-level taxa. BMC Genomics 2018; 19:931. [PMID: 30547746 PMCID: PMC6295017 DOI: 10.1186/s12864-018-5313-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/27/2018] [Indexed: 02/08/2023] Open
Abstract
Background Faecalibacterium prausnitzii is a ubiquitous member of the human gut microbiome, constituting up to 15% of the total bacteria in the human gut. Substantial evidence connects decreased levels of F. prausnitzii with the onset and progression of certain forms of inflammatory bowel disease, which has been attributed to its anti-inflammatory potential. Two phylogroups of F. prausnitzii have been identified, with a decrease in phylogroup I being a more sensitive marker of intestinal inflammation. Much of the genomic and physiological data available to date was collected using phylogroup II strains. Little analysis of F. prausnitzii genomes has been performed so far and genetic differences between phylogroups I and II are poorly understood. Results In this study we sequenced 11 additional F. prausnitzii genomes and performed comparative genomics to investigate intraspecies diversity, functional gene complement and the mobilome of 31 high-quality draft and complete genomes. We reveal a very low level of average nucleotide identity among F. prausnitzii genomes and a high level of genome plasticity. Two genomogroups can be separated based on differences in functional gene complement, albeit that this division does not fully agree with separation based on conserved gene phylogeny, highlighting the importance of horizontal gene transfer in shaping F. prausnitzii genomes. The difference between the two genomogroups is mainly in the complement of genes associated with catabolism of carbohydrates (such as a predicted sialidase gene in genomogroup I) and amino acids, as well as defense mechanisms. Conclusions Based on the combination of ANI of genomic sequences, phylogenetic analysis of core proteomes and functional differences we propose to separate the species F. prausnitzii into two new species level taxa: F. prausnitzii sensu stricto (neotype strain A2–165T = DSM 17677T = JCM 31915T) and F. moorei sp. nov. (type strain ATCC 27768T = NCIMB 13872T). Electronic supplementary material The online version of this article (10.1186/s12864-018-5313-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cormac Brian Fitzgerald
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | | | | | - Andrei V Chaplin
- Department of Microbiology and Virology, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland. .,School of Microbiology, University College Cork, Cork, Ireland.
| |
Collapse
|
1258
|
Sharp RC, Naser ES, Alcedo KP, Qasem A, Abdelli LS, Naser SA. Development of multiplex PCR and multi-color fluorescent in situ hybridization ( m-FISH) coupled protocol for detection and imaging of multi-pathogens involved in inflammatory bowel disease. Gut Pathog 2018; 10:51. [PMID: 30534203 PMCID: PMC6280354 DOI: 10.1186/s13099-018-0278-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/29/2018] [Indexed: 12/28/2022] Open
Abstract
Background Several pathogens have been debated to play a role in inflammatory bowel disease (IBD) including Crohn's disease (CD). None of these pathogens have been investigated together in same clinical samples. We developed a multiplex PCR and multi-color fluorescent in situ hybridization (m-FISH) protocols for simultaneous detection of CD-associated pathogens including Mycobacterium avium subspecies paratuberculosis (MAP), Klebsiella pneumoniae, and adherent-invasive Escherichia coli strain LF82. Methods The multiplex PCR is based on 1-h DNAzol® extraction protocol modified for rapid extraction of bacterial DNA from culture, blood, and intestinal biopsies. Oligonucleotide primers sequences unique to these pathogens were evaluated individually and in combinations using bioinformatics and experimental approaches. m-FISH was based on fluorescent-tagged oligonucleotides and confocal scanning laser microscopy (CSLM). Results Following several attempts, the concentration of the oligonucleotide primers and DNA templates and the PCR annealing temperatures were optimized. Multiplex PCR analyses revealed excellent amplification signal in trials where a single primer set and combinations of two and three primers sets were tested against a mixture of DNA from three different bacteria or a mixture of three bacterial cultures mixed in one tube before DNA extraction. Slides with individual and mixtures of bacterial cultures and intestinal tissue sections from IBD patients were tested by m-FISH and the CSLM images verified multiplex PCR results detected on 3% agarose gel. Conclusion We developed a 4-h multiplex PCR protocol, which was validated by m-FISH images, capable of detecting up to four genes from major pathogens associated with CD. The new protocol should serve as an excellent tool to support efforts to study multi-pathogens involved in CD and other autoimmune disease.
Collapse
Affiliation(s)
- Robert C Sharp
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Ebraheem S Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Karel P Alcedo
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Ahmad Qasem
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Latifa S Abdelli
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| | - Saleh A Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 4110 Libra Drive, Orlando, FL USA
| |
Collapse
|
1259
|
Causal Relationship between Diet-Induced Gut Microbiota Changes and Diabetes: A Novel Strategy to Transplant Faecalibacterium prausnitzii in Preventing Diabetes. Int J Mol Sci 2018; 19:ijms19123720. [PMID: 30467295 PMCID: PMC6320976 DOI: 10.3390/ijms19123720] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 11/18/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023] Open
Abstract
The incidence of metabolic disorders, including diabetes, has elevated exponentially during the last decades and enhanced the risk of a variety of complications, such as diabetes and cardiovascular diseases. In the present review, we have highlighted the new insights on the complex relationships between diet-induced modulation of gut microbiota and metabolic disorders, including diabetes. Literature from various library databases and electronic searches (ScienceDirect, PubMed, and Google Scholar) were randomly collected. There exists a complex relationship between diet and gut microbiota, which alters the energy balance, health impacts, and autoimmunity, further causes inflammation and metabolic dysfunction, including diabetes. Faecalibacterium prausnitzii is a butyrate-producing bacterium, which plays a vital role in diabetes. Transplantation of F. prausnitzii has been used as an intervention strategy to treat dysbiosis of the gut’s microbial community that is linked to the inflammation, which precedes autoimmune disease and diabetes. The review focuses on literature that highlights the benefits of the microbiota especially, the abundant of F. prausnitzii in protecting the gut microbiota pattern and its therapeutic potential against inflammation and diabetes.
Collapse
|
1260
|
Moen AEF, Lindstrøm JC, Tannæs TM, Vatn S, Ricanek P, Vatn MH, Jahnsen J. The prevalence and transcriptional activity of the mucosal microbiota of ulcerative colitis patients. Sci Rep 2018; 8:17278. [PMID: 30467421 PMCID: PMC6250705 DOI: 10.1038/s41598-018-35243-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/01/2018] [Indexed: 12/16/2022] Open
Abstract
Active microbes likely have larger impact on gut health status compared to inactive or dormant microbes. We investigate the composition of active and total mucosal microbiota of treatment-naïve ulcerative colitis (UC) patients to determine the microbial picture at the start-up phase of disease, using both a 16S rRNA transcript and gene amplicon sequencing. DNA and RNA were isolated from the same mucosal colonic biopsies. Our aim was to identify active microbial members of the microbiota in early stages of disease and reveal which members are present, but do not act as major players. We demonstrated differences in active and total microbiota of UC patients when comparing inflamed to non-inflamed tissue. Several taxa, among them the Proteobacteria phyla and families therein, revealed lower transcriptional activity despite a high presence. The Bifidobacteriaceae family of the Actinobacteria phylum showed lower abundance in the active microbiota, although no difference in presence was detected. The most abundant microbiota members of the inflamed tissue in UC patients were not the most active. Knowledge of active members of microbiota in UC patients could enhance our understanding of disease etiology. The active microbial community composition did not deviate from the total when comparing UC patients to non-IBD controls.
Collapse
Affiliation(s)
- Aina E Fossum Moen
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | - Jonas Christoffer Lindstrøm
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Tone Møller Tannæs
- Department of Clinical Molecular Biology (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway.
| | - Simen Vatn
- Department of Gastroenterology, Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | - Petr Ricanek
- Department of Gastroenterology, Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | - Morten H Vatn
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Jørgen Jahnsen
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Division of Medicine, Akershus University Hospital, Lørenskog, and University of Oslo, Oslo, Norway
| | | |
Collapse
|
1261
|
Mirsepasi-Lauridsen HC, Vrankx K, Engberg J, Friis-Møller A, Brynskov J, Nordgaard-Lassen I, Petersen AM, Krogfelt KA. Disease-Specific Enteric Microbiome Dysbiosis in Inflammatory Bowel Disease. Front Med (Lausanne) 2018; 5:304. [PMID: 30525037 PMCID: PMC6256240 DOI: 10.3389/fmed.2018.00304] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022] Open
Abstract
Inflammatory Bowel disease (IBD) is traditionally divided into Crohn's disease (CD) and ulcerative colitis (UC). UC is a relapsing non-transmural inflammatory disease that is restricted to the colon and is characterized by flare-ups of bloody diarrhea. CD is a chronic, segmental localized granulomatous disease that can affect any part of the entire gastrointestinal tract. Ileo-anal pouch is a procedure restoring functionality of the rectum after a colectomy. IBD is a multifactorial disease and flares of IBD are probably triggered by changes in the intestinal microbiota followed by an abnormal immune response. In this study, we aim to analyze the intestinal bacterial diversity in IBD patients during various stages of disease compared with healthy controls. Permission for human experiments and recruitment of participants was obtained from the Ethic Committee for Copenhagen County hospitals (Permission no. KA-03019, Permission no. KA-20060159). Stools from 26 healthy controls, 42 CD, 38 UC and 18 pouch patients were collected. Stool DNA extraction was performed using Qiagen, DNA mini stool kit Denmark. DGGE-PCR amplifying the V2-V3 region of 16S-rDNA gene of the bacteria was amplified by universal primers HDA1 and HDA2. Analysis of DGGE was performed blinded using BioNumerics version 7.5. After normalization, a DGGE gel band matching was performed. The similarities between profiles were calculated with a ranked Pearson correlation coefficient based on the band matching results using band intensities. Simpson's index of diversity and Pielou's species evenness were calculated. Based on the Shannon Diversity Index, UC patients had lower species diversity and bacterial evenness in comparison to healthy persons, p < 0.05. However, only CD and disease pouch patients had lower species diversity compared to those with inactive disease and healthy controls. Well-functioning pouch patients had decreased species evenness in comparison to diseased pouch patients and control group. During the active disease stage in CD and pouch, the patients have a low bacterial diversity in their gut when compared to the inactive stage. In UC patients, a generally low diversity was observed at all stages of the disease compared to healthy controls.
Collapse
Affiliation(s)
| | | | - Jørgen Engberg
- Department of Clinical Microbiology, Slagelse Hospital, Slagelse, Denmark
| | - Alice Friis-Møller
- Department of Clinical Microbiology, Hvidovre University Hospital, Hvidovre, Denmark
| | - Jørn Brynskov
- Department of Gastroenterology, Herlev University Hospital, Herlev, Denmark
| | | | - Andreas Munk Petersen
- Department of Clinical Microbiology, Hvidovre University Hospital, Hvidovre, Denmark.,Department of Gastroenterology, Hvidovre University Hospital, Hvidovre, Denmark
| | | |
Collapse
|
1262
|
Konieczna C, Słodziński M, Schmidt MT. Exopolysaccharides Produced by Lactobacillus rhamnosus KL 53A and Lactobacillus casei Fyos Affect Their Adhesion to Enterocytes. Pol J Microbiol 2018; 67:273-281. [PMID: 30451443 PMCID: PMC7256689 DOI: 10.21307/pjm-2018-032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2018] [Indexed: 11/11/2022] Open
Abstract
Probiotics promote and help to maintain beneficial microbiota composition of the gastrointestinal tract ecosystem and have a positive impact on the host’s health. Production of exopolysaccharides is an important feature of probiotic lactobacilli. It increases the chance of their survival in the gastrointestinal tract and promotes adhesion to the epithelium; therefore, exopolysaccharides are important for the process of colonization. Two lactic acid bacteria strains were used in this study: Lactobacillus rhamnosus KL 53A and Lactobacillus casei Fyos. Exopolysaccharides were isolated from bacterial cells and their monosaccharide composition was examined using liquid chromatography. The influence of exopolysaccharides on lactobacilli adhesion to enterocytes was studied after deglycosylation of the bacterial cells and incubation with the selected intestinal microbiota strains that metabolize polysaccharides – Faecalibacterium prausnitzii DSM 17677 and Blautia luti DSM 14534. Both deglycosylation and incubation with polysaccharide metabolizing strains influenced the ability of probiotic strains to adhere to enterocytes. Enzymatic deglycosylation decreased adhesion efficiency of L. rhamnosus KL 53A; however, co-incubation of both lactobacillus strains with F. prausnitzii DSM 17677 resulted in an increase of their adhesion efficiency. Exopolysaccharides are important adhesins of Lactobacillus spp. that influence their ability to colonize gut epithelium. Other members of gut microbiota can modify the adhesion property in situ; therefore the composition and metabolic state of commensal bacteria may influence their probiotic action.
Collapse
Affiliation(s)
- Corinna Konieczna
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, Poznan, Poland
| | - Michał Słodziński
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, Poznan, Poland
| | - Marcin T Schmidt
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, Poznan, Poland
| |
Collapse
|
1263
|
Moro Cantu-Jungles T, do Nascimento GE, Zhang X, Iacomini M, Cordeiro LMC, Hamaker BR. Soluble xyloglucan generates bigger bacterial community shifts than pectic polymers during in vitro fecal fermentation. Carbohydr Polym 2018; 206:389-395. [PMID: 30553337 DOI: 10.1016/j.carbpol.2018.11.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/22/2018] [Accepted: 11/06/2018] [Indexed: 11/30/2022]
Abstract
Xyloglucans and pectic polymers can be obtained from a variety of plants ubiquitous in the human diet, however, their fermentability in the colon and consequent nutritional benefits are poorly understood. Here, we evaluated metabolite profiles and bacterial shifts during in vitro fecal fermentations of two isolated pectic polymers and a xyloglucan. Depending on their chemical structure, pectic polymers were more acetogenic or propiogenic. Xyloglucan fermentation also resulted in elevated propionate if compared to FOS. Bacteroides plebeius, B. uniformis, Parabacteroides distasonis and bacterial groups such as Blautia, Lachnospira, Clostridiales and Lachnospiraceae, presented distinct abundances on each dietary fiber ferment. PCA and heat map analysis showed that major microbiota shifts occurred during xyloglucan fermentation, but not pectin fermentation. These data suggest that uncommon carbohydrate structures (i.e. isolated, soluble xyloglucan) in the diet hold the potential to generate larger shifts in microbiota communities than commonly consumed fibers (i.e. pectins).
Collapse
Affiliation(s)
- Thaisa Moro Cantu-Jungles
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Brazil; Whistler Center for Carbohydrate Research and Department of Food Science, Purdue University, West Lafayette, USA
| | | | - Xiaowei Zhang
- Whistler Center for Carbohydrate Research and Department of Food Science, Purdue University, West Lafayette, USA
| | - Marcello Iacomini
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Brazil
| | - Lucimara M C Cordeiro
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, Brazil.
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research and Department of Food Science, Purdue University, West Lafayette, USA.
| |
Collapse
|
1264
|
Yu LCH. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 2018; 25:79. [PMID: 30413188 PMCID: PMC6234774 DOI: 10.1186/s12929-018-0483-8] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease which arises as a result of the interaction of genetic, environmental, barrier and microbial factors leading to chronic inflammation in the intestine. Patients with IBD had a higher risk of developing colorectal carcinoma (CRC), of which the subset was classified as colitis-associated cancers. Genetic polymorphism of innate immune receptors had long been considered a major risk factor for IBD, and the mutations were also recently observed in CRC. Altered microbial composition (termed microbiota dybiosis) and dysfunctional gut barrier manifested by epithelial hyperpermeability and high amount of mucosa-associated bacteria were observed in IBD and CRC patients. The findings suggested that aberrant immune responses to penetrating commensal microbes may play key roles in fueling disease progression. Accumulative evidence demonstrated that mucosa-associated bacteria harbored colitogenic and protumoral properties in experimental models, supporting an active role of bacteria as pathobionts (commensal-derived opportunistic pathogens). Nevertheless, the host factors involved in bacterial dysbiosis and conversion mechanisms from lumen-dwelling commensals to mucosal pathobionts remain unclear. Based on the observation of gut leakiness in patients and the evidence of epithelial hyperpermeability prior to the onset of mucosal histopathology in colitic animals, it was postulated that the epithelial barrier dysfunction associated with mucosal enrichment of specific bacterial strains may predispose the shift to disease-associated microbiota. The speculation of leaky gut as an initiating factor for microbiota dysbiosis that eventually led to pathological consequences was proposed as the "common ground hypothesis", which will be highlighted in this review. Overall, the understanding of the core interplay between gut microbiota and epithelial barriers at early subclinical phases will shed light to novel therapeutic strategies to manage chronic inflammatory disorders and colitis-associated cancers.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Suite 1020, #1 Jen-Ai Rd. Sec. 1, Taipei, 100, Taiwan, Republic of China.
| |
Collapse
|
1265
|
Cetin-Karaca H, Morgan MC. Inactivation of Bacillus cereus spores in infant formula by combination of high pressure and trans-cinnamaldehyde. Lebensm Wiss Technol 2018. [DOI: 10.1016/j.lwt.2018.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1266
|
Dumas A, Bernard L, Poquet Y, Lugo-Villarino G, Neyrolles O. The role of the lung microbiota and the gut-lung axis in respiratory infectious diseases. Cell Microbiol 2018; 20:e12966. [PMID: 30329198 DOI: 10.1111/cmi.12966] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/05/2018] [Accepted: 10/14/2018] [Indexed: 12/13/2022]
Abstract
The pulmonary microbial community, described only a few years ago, forms a discreet part of the human host microbiota. The airway microbiota has been found to be substantially altered in the context of numerous respiratory disorders; nonetheless, its role in health and disease is as yet only poorly understood. Another important parameter to consider is the gut-lung axis, where distal (gut) immune modulation during respiratory disease is mediated by the gut microbiota. The use of specific microbiota strains, termed "probiotics," with beneficial effects on the host immunity and/or against pathogens, has proven successful in the treatment of intestinal disorders and is also showing promise in the context of airway diseases. In this review, we highlight the beneficial role of the body's commensal bacteria during airway infectious diseases, including recent evidence highlighting their local (lung) or distal (gut) contribution in this process.
Collapse
Affiliation(s)
- Alexia Dumas
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lucie Bernard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yannick Poquet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
1267
|
Khan S, Imran A, Malik A, Chaudhary AA, Rub A, Jan AT, Syed JB, Rolfo C. Bacterial imbalance and gut pathologies: Association and contribution of E. coli in inflammatory bowel disease. Crit Rev Clin Lab Sci 2018; 56:1-17. [DOI: 10.1080/10408363.2018.1517144] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shahanavaj Khan
- Nanomedicine Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Bioscience, Shri Ram Group of College (SRGC), Muzaffarnagar, India
| | - Ahamad Imran
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
| | - Abdul Malik
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Anis Ahmad Chaudhary
- Department of Pharmacology, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Abdur Rub
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | - Arif Tasleem Jan
- School of Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Jakeera Begum Syed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
- College of Medicine and Dentistry, Dar Al Uloom University, Riyadh, Saudi Arabia
| | - Christian Rolfo
- Phase I-Early Clinical Trials Unit, Oncology Department and Multidisciplinary Oncology Center Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
1268
|
Sayers E, MacGregor A, Carding SR. Drug-microbiota interactions and treatment response: Relevance to rheumatoid arthritis. AIMS Microbiol 2018; 4:642-654. [PMID: 31294239 PMCID: PMC6613334 DOI: 10.3934/microbiol.2018.4.642] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/24/2018] [Indexed: 01/16/2023] Open
Abstract
Knowledge about associations between changes in the structure and/or function of intestinal microbes (the microbiota) and the pathogenesis of various diseases is expanding. However, interactions between the intestinal microbiota and different pharmaceuticals and the impact of these on responses to treatment are less well studied. Several mechanisms are known by which drug-microbiota interactions can influence drug bioavailability, efficacy, and/or toxicity. This includes direct activation or inactivation of drugs by microbial enzymes which can enhance or reduce drug effectiveness. The extensive metabolic capabilities of the intestinal microbiota make it a hotspot for drug modification. However, drugs can also influence the microbiota profoundly and change the outcome of interactions with the host. Additionally, individual microbiota signatures are unique, leading to substantial variation in host responses to particular drugs. In this review, we describe several known and emerging examples of how drug-microbiota interactions influence the responses of patients to treatment for various diseases, including inflammatory bowel disease, type 2 diabetes and cancer. Focussing on rheumatoid arthritis (RA), a chronic inflammatory disease of the joints which has been linked with microbial dysbiosis, we propose mechanisms by which the intestinal microbiota may affect responses to treatment with methotrexate which are highly variable. Furthering our knowledge of this subject will eventually lead to the adoption of new treatment strategies incorporating microbiota signatures to predict or improve treatment outcomes.
Collapse
Affiliation(s)
- Ellie Sayers
- Gut Health and Food Safety Programme, Quadram Institute Bioscience, Norwich, Norfolk, NR4 7UA, UK.,Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| | - Alex MacGregor
- Gut Health and Food Safety Programme, Quadram Institute Bioscience, Norwich, Norfolk, NR4 7UA, UK
| | - Simon R Carding
- Gut Health and Food Safety Programme, Quadram Institute Bioscience, Norwich, Norfolk, NR4 7UA, UK.,Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, Norfolk, NR4 7TJ, UK
| |
Collapse
|
1269
|
Al Bakir I, Curtius K, Graham TA. From Colitis to Cancer: An Evolutionary Trajectory That Merges Maths and Biology. Front Immunol 2018; 9:2368. [PMID: 30386335 PMCID: PMC6198656 DOI: 10.3389/fimmu.2018.02368] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 09/24/2018] [Indexed: 12/25/2022] Open
Abstract
Patients with inflammatory bowel disease have an increased risk of developing colorectal cancer, and this risk is related to disease duration, extent, and cumulative inflammation burden. Carcinogenesis follows the principles of Darwinian evolution, whereby somatic cells acquire genomic alterations that provide them with a survival and/or growth advantage. Colitis represents a unique situation whereby routine surveillance endoscopy provides a serendipitous opportunity to observe somatic evolution over space and time in vivo in a human organ. Moreover, somatic evolution in colitis is evolution in the ‘fast lane': the repeated rounds of inflammation and mucosal healing that are characteristic of the disease accelerate the evolutionary process and likely provide a strong selective pressure for inflammation-adapted phenotypic traits. In this review, we discuss the evolutionary dynamics of pre-neoplastic clones in colitis with a focus on how measuring their evolutionary trajectories could deliver a powerful way to predict future cancer occurrence. Measurements of somatic evolution require an interdisciplinary approach that combines quantitative measurement of the genotype, phenotype and the microenvironment of somatic cells–paying particular attention to spatial heterogeneity across the colon–together with mathematical modeling to interpret these data within an evolutionary framework. Here we take a practical approach in discussing how and why the different “evolutionary ingredients” can and should be measured, together with our viewpoint on subsequent translation into clinical practice. We highlight the open questions in the evolution of colitis-associated cancer as a stimulus for future work.
Collapse
Affiliation(s)
- Ibrahim Al Bakir
- Evolution and Cancer Laboratory, Centre for Tumour Biology, Barts Cancer Institute, London, United Kingdom.,Inflammatory Bowel Disease Unit, St Mark's Hospital, Harrow, United Kingdom
| | - Kit Curtius
- Evolution and Cancer Laboratory, Centre for Tumour Biology, Barts Cancer Institute, London, United Kingdom
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Centre for Tumour Biology, Barts Cancer Institute, London, United Kingdom
| |
Collapse
|
1270
|
Brusaferro A, Cavalli E, Farinelli E, Cozzali R, Principi N, Esposito S. Gut dysbiosis and paediatric Crohn's disease. J Infect 2018; 78:1-7. [PMID: 30336176 DOI: 10.1016/j.jinf.2018.10.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/24/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The main objective of this manuscript is to discuss our present knowledge of the relationships between dysbiosis and paediatric Crohn's disease (CD). The therapeutic role of the methods currently used to re-establish normal gut microbiota composition is also analysed. METHODS PubMed was used to search for all of the studies published from January 2008 to June 2018 using the key words: "Crohn's disease" and "gut dysbiosis" or "microbiota" or "microbioma" or "probiotic" and "children" or "paediatric". More than 100 articles were found, but only those published in English or providing evidence-based data were included in the evaluation. RESULTS Gut microbiota are primary actors in CD's pathogenesis. The new techniques developed in metagenomics allow us to reveal new details of microbiota composition in healthy subjects and CD patients, and to elucidate the link between microbiota and numerous pathologies, such as obesity, allergies and type 1 diabetes mellitus. CONCLUSION Discoveries on the role of gut microbiota could potentially disclose new therapeutic options for CD treatment and improve the existing therapies. Further studies are needed to facilitate the diagnosis and tailor the therapy of a pathology that is an increasing burden on public health.
Collapse
Affiliation(s)
- Andrea Brusaferro
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, Perugia 06129, Italy
| | - Elena Cavalli
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, Perugia 06129, Italy
| | - Edoardo Farinelli
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, Perugia 06129, Italy
| | - Rita Cozzali
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, Perugia 06129, Italy
| | | | - Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, Perugia 06129, Italy.
| |
Collapse
|
1271
|
Zhu Q, Gao R, Zhang Y, Pan D, Zhu Y, Zhang X, Yang R, Jiang R, Xu Y, Qin H. Dysbiosis signatures of gut microbiota in coronary artery disease. Physiol Genomics 2018; 50:893-903. [PMID: 30192713 DOI: 10.1152/physiolgenomics.00070.2018] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota dysbiosis has been considered to be an important risk factor that contributes to coronary artery disease (CAD), but limited evidence exists about the involvement of gut microbiota in the disease. Our study aimed to characterize the dysbiosis signatures of gut microbiota in coronary artery disease. The gut microbiota represented in stool samples were collected from 70 patients with coronary artery disease and 98 healthy controls. 16S rRNA sequencing was applied, and bioinformatics methods were used to decipher taxon signatures and function alteration, as well as the microbial network and diagnostic model of gut microbiota in coronary artery disease. Gut microbiota showed decreased diversity and richness in patients with coronary artery disease. The composition of the microbial community changed; Escherichia-Shigella [false discovery rate (FDR = 7.5*10−5] and Enterococcus (FDR = 2.08*10−7) were significant enriched, while Faecalibacterium (FDR = 6.19*10−10), Subdoligranulum (FDR = 1.63*10−6), Roseburia (FDR = 1.95*10−9), and Eubacterium rectale (FDR = 2.35*10−4) were significant depleted in the CAD group. Consistent with the taxon changes, functions such as amino acid metabolism, phosphotransferase system, propanoate metabolism, lipopolysaccharide biosynthesis, and protein and tryptophan metabolism were found to be enhanced in CAD patients. The microbial network revealed that Faecalibacterium and Escherichia-Shigella were the microbiotas that dominated in the healthy control and CAD groups, respectively. The microbial diagnostic model based on random forest also showed probability in identifying those who suffered from CAD. Our study successfully identifies the dysbiosis signature, dysfunctions, and comprehensive networks of gut microbiota in CAD patients. Thus, modulation targeting the gut microbiota may be a novel strategy for CAD treatment.
Collapse
Affiliation(s)
- Qi Zhu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Renyuan Gao
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Yi Zhang
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dengdeng Pan
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Yefei Zhu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Xiaohui Zhang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Rong Yang
- Department of Pediatrics, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rong Jiang
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huanlong Qin
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
1272
|
Liu B, Wang W, Zhu X, Sun X, Xiao J, Li D, Cui Y, Wang C, Shi Y. Response of Gut Microbiota to Dietary Fiber and Metabolic Interaction With SCFAs in Piglets. Front Microbiol 2018; 9:2344. [PMID: 30323803 PMCID: PMC6172335 DOI: 10.3389/fmicb.2018.02344] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/12/2018] [Indexed: 02/02/2023] Open
Abstract
Dietary fiber (DF) is increasingly thought to regulate diversity of piglet gut microbiota to alleviate weaning stress in piglets. This study was conducted to investigate the effects of DF on growth performance of piglets and composition of their gut microbiota, as well as the interaction between gut microbiota and short-chain fatty acids (SCFAs) in piglets. A total of 840 piglets were allocated to three dietary treatments consisting of a control group (CG), an alfalfa meal group (AG), and a commodity concentrated fiber group (OG) in a 30-day feeding trial. Gut mucosa and feces samples were used to determine bacterial community diversity by 16S rRNA gene amplicon sequencing. Fiber treatment had a positive effect on growth performance and metabolism of SCFAs in piglets, in particular, compared with CG, the diarrhea rate was significantly decreased, and the content of propionic acid (PA) in the cecum was markedly increased in AG. The Shannon indices of the jejunum microbiota in AG were higher than CG. At the genus level, compared to CG, in the duodenum, the relative abundance of Paenibacillus in AG and OG was higher; in the jejunum, the relative abundances of Bacillus, Oceanobacillus, Paenibacillus, Lactococcus, Enterococcus, and Exiguobacterium were higher, whereas the relative abundance of Mycoplasma was lower in AG; in the cecum, there was also lower relative abundance of Helicobacter in AG and OG, and furthermore, the relative abundance of Faecalibacterium in OG was higher than in CG and AG. Spearman correlation analysis showed that Pseudobutyrivibrio was positively correlated with acetic acid, PA, and butyric acid (BA), while Bacteroides and Anaerotruncus were negatively correlated with PA and BA. In addition, microbiota analyses among different intestine segments showed distinct differences in microbiota between the proximal and distal intestines. Bacteria in the proximal segments were mainly Firmicutes, while bacteria in the distal segments were mainly Bacteroidetes and Firmicutes. Overall, these findings suggested that DF treatment could reduce the diarrhea rate of piglets and had beneficial effects on gut health, which might be attributed to the alteration in gut microbiota induced by DF and the interaction of the gut microbiota with SCFAs.
Collapse
Affiliation(s)
- Boshuai Liu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Wenjing Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xiaoyan Zhu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
| | - Xiao Sun
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Junnan Xiao
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Defeng Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
| | - Yalei Cui
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
| | - Chengzhang Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
| | - Yinghua Shi
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China.,Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China
| |
Collapse
|
1273
|
Pirone-Davies C, Chen Y, Pightling A, Ryan G, Wang Y, Yao K, Hoffmann M, Allard MW. Genes significantly associated with lineage II food isolates of Listeria monocytogenes. BMC Genomics 2018; 19:708. [PMID: 30253738 PMCID: PMC6157050 DOI: 10.1186/s12864-018-5074-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 09/12/2018] [Indexed: 01/02/2023] Open
Abstract
Background Listeria monocytogenes is a widespread foodborne pathogen that can cause listeriosis, a potentially fatal infection. L. monocytogenes is subdivided into four phylogenetic lineages, with the highest incidence of listeriosis occurring within lineage I followed by lineage II. Strains of L. monocytogenes differ in their phenotypic characteristics, including virulence. However, the genetic bases for these observed differences are not well understood, and current efforts to monitor L. monocytogenes in food consider all strains to be equally virulent. We use a comparative genomics approach to identify genes and single nucleotide polymorphisms (SNPs) in 174 clinical and food isolates of L. monocytogenes that potentially contribute to virulence or the capacity to adapt to food environments. Results No SNPs are significantly associated with food or clinical isolates. No genes are significantly associated with food or clinical isolates from lineage I, but eight genes consisting of multiple homologues are associated with lineage II food isolates. These include three genes which encode hypothetical proteins, the cadmium resistance genes cadA and cadC, the multi-drug resistance gene ebrB, a quaternary ammonium compound resistance gene qac, and a regulatory gene. All eight genes are plasmid-borne, and most closed L. monocytogenes plasmids carry at least five of the genes (24/27). In addition, plasmids are more frequently associated with lineage II food isolates than with lineage II clinical isolates. Conclusions We identify eight genes that are significantly associated with food isolates in lineage II. Interestingly, the eight genes are virtually absent in lineage II outbreak isolates, are composed of homologues which show a nonrandom distribution among lineage I serotypes, and the sequences are highly conserved across 27 closed Listeria plasmids. The functions of these genes should be explored further and will contribute to our understanding of how L. monocytogenes adapts to the host and food environments. Moreover, these genes may also be useful as markers for risk assessment models of either pathogenicity or the ability to proliferate in food and the food processing environment. Electronic supplementary material The online version of this article (10.1186/s12864-018-5074-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cary Pirone-Davies
- Division of Microbiology, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA.
| | - Yi Chen
- Division of Microbiology, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA
| | - Arthur Pightling
- Office of Analytics and Outreach, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA
| | - Gina Ryan
- Division of Microbiology, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA
| | - Yu Wang
- Office of Analytics and Outreach, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA
| | - Kuan Yao
- Division of Microbiology, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA
| | - Maria Hoffmann
- Division of Microbiology, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA
| | - Marc W Allard
- Division of Microbiology, Office of Regulatory Science, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, USA
| |
Collapse
|
1274
|
Michelini S, Balakrishnan B, Parolo S, Matone A, Mullaney JA, Young W, Gasser O, Wall C, Priami C, Lombardo R, Kussmann M. A reverse metabolic approach to weaning: in silico identification of immune-beneficial infant gut bacteria, mining their metabolism for prebiotic feeds and sourcing these feeds in the natural product space. MICROBIOME 2018; 6:171. [PMID: 30241567 PMCID: PMC6151060 DOI: 10.1186/s40168-018-0545-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/30/2018] [Indexed: 05/13/2023]
Abstract
BACKGROUND Weaning is a period of marked physiological change. The introduction of solid foods and the changes in milk consumption are accompanied by significant gastrointestinal, immune, developmental, and microbial adaptations. Defining a reduced number of infections as the desired health benefit for infants around weaning, we identified in silico (i.e., by advanced public domain mining) infant gut microbes as potential deliverers of this benefit. We then investigated the requirements of these bacteria for exogenous metabolites as potential prebiotic feeds that were subsequently searched for in the natural product space. RESULTS Using public domain literature mining and an in silico reverse metabolic approach, we constructed probiotic-prebiotic-food associations, which can guide targeted feeding of immune health-beneficial microbes by weaning food; analyzed competition and synergy for (prebiotic) nutrients between selected microbes; and translated this information into designing an experimental complementary feed for infants enrolled in a pilot clinical trial ( http://www.nourishtoflourish.auckland.ac.nz/ ). CONCLUSIONS In this study, we applied a benefit-oriented microbiome research strategy for enhanced early-life immune health. We extended from "classical" to molecular nutrition aiming to identify nutrients, bacteria, and mechanisms that point towards targeted feeding to improve immune health in infants around weaning. Here, we present the systems biology-based approach we used to inform us on the most promising prebiotic combinations known to support growth of beneficial gut bacteria ("probiotics") in the infant gut, thereby favorably promoting development of the immune system.
Collapse
Affiliation(s)
- Samanta Michelini
- The Microsoft Research–University of Trento Centre for Computational and Systems Biology, Rovereto, Italy
| | - Biju Balakrishnan
- The Liggins Institute, the University of Auckland, Auckland, New Zealand
| | - Silvia Parolo
- The Microsoft Research–University of Trento Centre for Computational and Systems Biology, Rovereto, Italy
| | - Alice Matone
- The Microsoft Research–University of Trento Centre for Computational and Systems Biology, Rovereto, Italy
| | - Jane A. Mullaney
- AgResearch, Food & Bio-based Products, Palmerston North, New Zealand
- Riddet Institute, Palmerston North, New Zealand
| | - Wayne Young
- AgResearch, Food & Bio-based Products, Palmerston North, New Zealand
- Riddet Institute, Palmerston North, New Zealand
| | - Olivier Gasser
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Clare Wall
- Discipline of Nutrition, School of Medical Science, University of Auckland, Auckland, New Zealand
| | - Corrado Priami
- The Microsoft Research–University of Trento Centre for Computational and Systems Biology, Rovereto, Italy
- Department of Computer Science, University of Pisa, Pisa, Italy
| | - Rosario Lombardo
- The Microsoft Research–University of Trento Centre for Computational and Systems Biology, Rovereto, Italy
| | - Martin Kussmann
- The Liggins Institute, the University of Auckland, Auckland, New Zealand
- National Science Challenge “High Value Nutrition”, Auckland, New Zealand
| |
Collapse
|
1275
|
Wang Q, Li F, Liang B, Liang Y, Chen S, Mo X, Ju Y, Zhao H, Jia H, Spector TD, Xie H, Guo R. A metagenome-wide association study of gut microbiota in asthma in UK adults. BMC Microbiol 2018; 18:114. [PMID: 30208875 PMCID: PMC6134768 DOI: 10.1186/s12866-018-1257-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 09/04/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Asthma, one of the most common chronic respiratory disorders, is associated with the hyper-activation of the T-cell subset of adaptive immunity. The gut microbiota may be involved in the development of asthma through the production of short-chain fatty acids (SCFAs), exhibiting modulatory effects on Th. So, we performed a metagenome-wide association study (MWAS) of the fecal microbiota from individuals with asthma and healthy controls. And that was the first case to resolve the relationship between asthma and microbiome among UK adults. RESULTS The microbiota of the individuals with asthma consisted of fewer microbial entities than the microbiota of healthy individuals. Faecalibacterium prausnitzii, Sutterella wadsworthensis and Bacteroides stercoris were depleted in cases, whereas Clostridiums with Eggerthella lenta were over-represented in individuals with asthma. Functional analysis shows that the SCFAs might be altered in the microbiota of asthma patients. CONCLUSION In all, the adult human gut microbiome of asthma patients is clearly different from healthy controls. The functional and taxa results showed that the change of asthma patients might related to SCFAs.
Collapse
Affiliation(s)
- Qi Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China.,BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Fei Li
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China.,BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Bishan Liang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Yuhu Liang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China.,BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Sijie Chen
- BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Xiaodong Mo
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China.,BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yanmei Ju
- BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Hui Zhao
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, 518083, China.,BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Huijue Jia
- BGI-Shenzhen, Shenzhen, 518083, China.,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China.,Macau University of Science and Technology, Taipa, Macau, 999078, China.,Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen, 518083, China
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, SE1 7EH, UK
| | | | - Ruijin Guo
- BGI-Shenzhen, Shenzhen, 518083, China. .,China National Genebank, BGI-Shenzhen, Shenzhen, 518083, China. .,Macau University of Science and Technology, Taipa, Macau, 999078, China.
| |
Collapse
|
1276
|
Lopez-Siles M, Enrich-Capó N, Aldeguer X, Sabat-Mir M, Duncan SH, Garcia-Gil LJ, Martinez-Medina M. Alterations in the Abundance and Co-occurrence of Akkermansia muciniphila and Faecalibacterium prausnitzii in the Colonic Mucosa of Inflammatory Bowel Disease Subjects. Front Cell Infect Microbiol 2018; 8:281. [PMID: 30245977 PMCID: PMC6137959 DOI: 10.3389/fcimb.2018.00281] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/25/2018] [Indexed: 12/21/2022] Open
Abstract
Akkermansia muciniphila and Faecalibacterium prausnitzii, cohabitants in the intestinal mucosa, are considered members of a healthy microbiota and reduction of both species occurs in several intestinal disorders, including inflammatory bowel disease. Little is known however about a possible link between the reduction in quantity of these species, and in which circumstances this may occur. This study aims to determine the abundances and co-occurrence of the two species in order to elucidate conditions that may compromise their presence in the gut. Loads of A. muciniphila, total F. prausnitzii and its two phylogroup (16S rRNA gene copies) were determined by quantitative polymerase chain reaction in colonic biopsies from 17 healthy controls (H), 23 patients with ulcerative colitis (UC), 31 patients with Crohn's disease (CD), 3 with irritable bowel syndrome (IBS) and 3 with colorectal cancer (CRC). Data were normalized to total bacterial 16S rRNA gene copies in the same sample. Prevalence, relative abundances and correlation analyses were performed according to type of disease and considering relevant clinical characteristics of patients such as IBD location, age of disease onset, CD behavior, current medication and activity status. Co-occurrence of both species was found in 29% of H, 65% of UC and 29% of CD. Lower levels of total F. prausnitzii and phylogroups were found in subjects with CD, compared with H subjects (P ≤ 0.044). In contrast, no differences were found with the regard to A. muciniphila abundance across different disease states, but CD patients with disease onset below 16 years of age featured a marked depletion of this species. In CD patients, correlation between A. muciniphila and total F. prausnitzii (ρ = 0.362, P = 0.045) was observed, and particularly in those with non-stricturing, non-penetrating disease behavior and under moderate immunosuppressants therapy. Altogether, this study revealed that co-occurrence of both species differs between disease status. In addition, IBD patients featured a reduction of F. prausnitzii but similar loads of A. muciniphila when compared to H subjects, with the exception of those with early onset CD. Depletion of A. muciniphila in this subgroup of subjects suggests that it could be a potential biomarker to assist in pediatric CD diagnosis.
Collapse
Affiliation(s)
- Mireia Lopez-Siles
- Laboratory of Molecular Microbiology, Biology Department, Universitat de Girona, Girona, Spain
| | - Núria Enrich-Capó
- Laboratory of Molecular Microbiology, Biology Department, Universitat de Girona, Girona, Spain
| | - Xavier Aldeguer
- Department of Gastroenterology, Hospital Dr. Josep Trueta, Girona, Spain
| | - Miriam Sabat-Mir
- Department of Gastroenterology, Hospital Santa Caterina, Girona, Spain
| | - Sylvia H. Duncan
- Microbiology Group, Rowett Institute of Nutrition and Health, Aberdeen, United Kingdom
| | - L. Jesús Garcia-Gil
- Laboratory of Molecular Microbiology, Biology Department, Universitat de Girona, Girona, Spain
| | | |
Collapse
|
1277
|
Gîlcă-Blanariu GE, Diaconescu S, Ciocoiu M, Ștefănescu G. New Insights into the Role of Trace Elements in IBD. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1813047. [PMID: 30258848 PMCID: PMC6146599 DOI: 10.1155/2018/1813047] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/11/2018] [Accepted: 08/11/2018] [Indexed: 02/07/2023]
Abstract
Micronutrient deficiencies are common in inflammatory bowel disease and have clinical impact, being both a sign of complicated disease and a cause of morbidity. The involved systemic inflammatory response is responsible for altering the concentration of a wide range of trace elements in the serum, including zinc and selenium. This review summarizes recent advances and evidence-based knowledge regarding the impact of selenium and zinc on oxidative stress and microbiota changes in IBD patients. Getting new insight into the impact of malnutrition, particularly on the micronutrients' impact on the development, composition, and metabolism of microbiota, as well as the influence of oxidative stress and the mucosal immune response, could help in implementing new management strategies for IBD patients, with focus on a more integrated approach.
Collapse
Affiliation(s)
| | - Smaranda Diaconescu
- Department of Pediatrics, Titu Maiorescu University, Faculty of Medicine, Bucharest, Romania
| | - Manuela Ciocoiu
- Department of Pathophysiology, Grigore T Popa University of Medicine and Pharmacy, Iași, Romania
| | - Gabriela Ștefănescu
- Department of Gastroenterology, Grigore T Popa University of Medicine and Pharmacy, Iași, Romania
| |
Collapse
|
1278
|
Alemán JO, Bokulich NA, Swann JR, Walker JM, De Rosa JC, Battaglia T, Costabile A, Pechlivanis A, Liang Y, Breslow JL, Blaser MJ, Holt PR. Fecal microbiota and bile acid interactions with systemic and adipose tissue metabolism in diet-induced weight loss of obese postmenopausal women. J Transl Med 2018; 16:244. [PMID: 30176893 PMCID: PMC6122649 DOI: 10.1186/s12967-018-1619-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 08/25/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Microbiota and bile acids in the gastrointestinal tract profoundly alter systemic metabolic processes. In obese subjects, gradual weight loss ameliorates adipose tissue inflammation and related systemic changes. We assessed how rapid weight loss due to a very low calorie diet (VLCD) affects the fecal microbiome and fecal bile acid composition, and their interactions with the plasma metabolome and subcutaneous adipose tissue inflammation in obesity. METHODS We performed a prospective cohort study of VLCD-induced weight loss of 10% in ten grades 2-3 obese postmenopausal women in a metabolic unit. Baseline and post weight loss evaluation included fasting plasma analyzed by mass spectrometry, adipose tissue transcription by RNA sequencing, stool 16S rRNA sequencing for fecal microbiota, fecal bile acids by mass spectrometry, and urinary metabolic phenotyping by 1H-NMR spectroscopy. Outcome measures included mixed model correlations between changes in fecal microbiota and bile acid composition with changes in plasma metabolite and adipose tissue gene expression pathways. RESULTS Alterations in the urinary metabolic phenotype following VLCD-induced weight loss were consistent with starvation ketosis, protein sparing, and disruptions to the functional status of the gut microbiota. We show that the core microbiome was preserved during VLCD-induced weight loss, but with changes in several groups of bacterial taxa with functional implications. UniFrac analysis showed overall parallel shifts in community structure, corresponding to reduced abundance of the genus Roseburia and increased Christensenellaceae;g__ (unknown genus). Imputed microbial functions showed changes in fat and carbohydrate metabolism. A significant fall in fecal total bile acid concentration and reduced deconjugation and 7-α-dihydroxylation were accompanied by significant changes in several bacterial taxa. Individual bile acids in feces correlated with amino acid, purine, and lipid metabolic pathways in plasma. Furthermore, several fecal bile acids and bacterial species correlated with altered gene expression pathways in adipose tissue. CONCLUSIONS VLCD dietary intervention in obese women changed the composition of several fecal microbial populations while preserving the core fecal microbiome. Changes in individual microbial taxa and their functions correlated with variations in the plasma metabolome, fecal bile acid composition, and adipose tissue transcriptome. Trial Registration ClinicalTrials.gov NCT01699906, 4-Oct-2012, Retrospectively registered. URL- https://clinicaltrials.gov/ct2/show/NCT01699906.
Collapse
Affiliation(s)
- José O. Alemán
- Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
- New York University Langone Medical Center, 423 East 23rd St, New York, NY 10016 USA
| | - Nicholas A. Bokulich
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ USA
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, S1W7 2AZ UK
| | - Jeanne M. Walker
- Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | | | - Thomas Battaglia
- New York University Langone Medical Center, 423 East 23rd St, New York, NY 10016 USA
| | - Adele Costabile
- Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Reading, RG6 6AP UK
| | - Alexandros Pechlivanis
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, S1W7 2AZ UK
| | - Yupu Liang
- Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Jan L. Breslow
- Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Martin J. Blaser
- New York University Langone Medical Center, 423 East 23rd St, New York, NY 10016 USA
| | - Peter R. Holt
- Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| |
Collapse
|
1279
|
Nusbaum DJ, Sun F, Ren J, Zhu Z, Ramsy N, Pervolarakis N, Kunde S, England W, Gao B, Fiehn O, Michail S, Whiteson K. Gut microbial and metabolomic profiles after fecal microbiota transplantation in pediatric ulcerative colitis patients. FEMS Microbiol Ecol 2018; 94:5053801. [PMID: 30010747 PMCID: PMC6454419 DOI: 10.1093/femsec/fiy133] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022] Open
Abstract
Ulcerative colitis is a chronic inflammatory disease of the colon that carries a significant disease burden in children. Therefore, new therapeutic approaches are being explored to help children living with this disease. Fecal microbiota transplantation (FMT) has been successful in some children with ulcerative colitis. However, the mechanism of its therapeutic effect in this patient population is not well understood. To characterize changes in gut microbial and metabolomic profiles after FMT, we performed 16S rRNA gene sequencing, shotgun metagenomic sequencing, virome analysis and untargeted metabolomics by gas chromatography-time of flight-mass spectrometry on stool samples collected before and after FMT from four children with ulcerative colitis who responded to this treatment. Alpha diversity of the gut microbiota increased after intervention, with species richness rising from 251 (S.D. 125) to 358 (S.D. 27). In responders, the mean relative abundance of bacteria in the class Clostridia shifted toward donor levels, increasing from 33% (S.D. 11%) to 54% (S.D. 16%). Patient metabolomic and viromic profiles exhibited a similar but less pronounced shift toward donor profiles after FMT. The fecal concentrations of several metabolites were altered after FMT, correlating with clinical improvement. Larger studies using a similar multi-omics approach may suggest novel strategies for the treatment of pediatric ulcerative colitis.
Collapse
Affiliation(s)
- David J Nusbaum
- Keck School of Medicine of the University of Southern California, Department of Pediatrics, Los Angeles, CA, USA 1975 Zonal Ave, Los Angeles, CA, USA 90033
| | - Fengzhu Sun
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA 1050 Childs Way, RRI201, Los Angeles, CA, USA 90089
| | - Jie Ren
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA 1050 Childs Way, RRI201, Los Angeles, CA, USA 90089
| | - Zifan Zhu
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA 1050 Childs Way, RRI201, Los Angeles, CA, USA 90089
| | - Natalie Ramsy
- Keck School of Medicine of the University of Southern California, Department of Pediatrics, Los Angeles, CA, USA 1975 Zonal Ave, Los Angeles, CA, USA 90033
| | - Nicholas Pervolarakis
- Center for Complex Biological Systems, University of California, Irvine, CA, USA 2620 Biological Sciences III, Irvine, CA, USA 92697
| | - Sachin Kunde
- Helen DeVos Children's Hospital, Grand Rapids, MI, USA 35 Michigan St NE, Ste 4150, Grand Rapids, MI, USA 49503
| | - Whitney England
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA 3315 McGaugh Hall, Irvine, CA, USA 92697
| | - Bei Gao
- West Coast Metabolomics Center, University of California, Davis, CA, USA 451 Health Sciences Drive, Davis, CA, USA 95616
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, CA, USA 451 Health Sciences Drive, Davis, CA, USA 95616
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia P.O. Box 80203, Jeddah, Saudi Arabia 21589
| | - Sonia Michail
- Keck School of Medicine of the University of Southern California, Department of Pediatrics, Los Angeles, CA, USA 1975 Zonal Ave, Los Angeles, CA, USA 90033
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA 3315 McGaugh Hall, Irvine, CA, USA 92697
| |
Collapse
|
1280
|
Gurry T, Gibbons SM, Nguyen LTT, Kearney SM, Ananthakrishnan A, Jiang X, Duvallet C, Kassam Z, Alm EJ. Predictability and persistence of prebiotic dietary supplementation in a healthy human cohort. Sci Rep 2018; 8:12699. [PMID: 30139999 PMCID: PMC6107591 DOI: 10.1038/s41598-018-30783-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/26/2018] [Indexed: 01/05/2023] Open
Abstract
Dietary interventions to manipulate the human gut microbiome for improved health have received increasing attention. However, their design has been limited by a lack of understanding of the quantitative impact of diet on a host’s microbiota. We present a highly controlled diet perturbation experiment in a healthy, human cohort in which individual micronutrients are spiked in against a standardized background. We identify strong and predictable responses of specific microbes across participants consuming prebiotic spike-ins, at the level of both strains and functional genes, suggesting fine-scale resource partitioning in the human gut. No predictable responses to non-prebiotic micronutrients were found. Surprisingly, we did not observe decreases in day-to-day variability of the microbiota compared to a complex, varying diet, and instead found evidence of diet-induced stress and an associated loss of biodiversity. Our data offer insights into the effect of a low complexity diet on the gut microbiome, and suggest that effective personalized dietary interventions will rely on functional, strain-level characterization of a patient’s microbiota.
Collapse
Affiliation(s)
- Thomas Gurry
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | | | - Sean M Gibbons
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Le Thanh Tu Nguyen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Sean M Kearney
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ashwin Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Xiaofang Jiang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Claire Duvallet
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Zain Kassam
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,OpenBiome, Somerville, MA, 02143, USA
| | - Eric J Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA. .,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA. .,The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
1281
|
Jin DX, Zou HW, Liu SQ, Wang LZ, Xue B, Wu D, Tian G, Cai J, Yan TH, Wang ZS, Peng QH. The underlying microbial mechanism of epizootic rabbit enteropathy triggered by a low fiber diet. Sci Rep 2018; 8:12489. [PMID: 30131509 PMCID: PMC6104036 DOI: 10.1038/s41598-018-30178-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023] Open
Abstract
Epizootic rabbit enteropathy (ERE) is reproduced successfully in the present study by feeding rabbits a low-fibre diet, and high-throughput sequencing and quantitative real-time PCR (qPCR) analysis were applied to examine the microbial variations in the stomach, small intestine and caecum. The evenness was disturbed and the richness was decreased in the ERE groups. When the rabbits were suffering from ERE, the abundance of the Firmicutes was decreased in three parts of the digestive tract, whereas the Proteobacteria was increased in the stomach and caecum, the Bacteroidetes and Verrucomicrobia were increased in the small intestine. Correlation analysis showed that the reduced concentrations of TVFA and butyrate in the caeca of the ERE group were attributed to the decreased abundances of genera such as Lactobacillus, Alistipes and other fibrolytic bacteria and butyrate- producing bacteria such as Eubacterium and Faecalibacterium. It is concluded that, in terms of microorganisms, the overgrowth of Bacteroides fragilis, Clostridium perfringen, Enterobacter sakazakii and Akkermansia muciniphila and inhibition of Bifidobacterium spp. and Butyrivibrio fibrisolvens in the stomach, small intestine and caecum resulted in a decrease in butyrate yield, leading to the incidence of ERE, and the probability of developing ERE could be manipulated by adjusting the dietary fibre level.
Collapse
Affiliation(s)
- Ding Xing Jin
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - Hua Wei Zou
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - Si Qiang Liu
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - Li Zhi Wang
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - Bai Xue
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - De Wu
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - Gang Tian
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - Jingyi Cai
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China
| | - Tian Hai Yan
- Agri-Food and Biosciences Institute, Hillsborough, Co. Down, BT26 6DR, United Kingdom
| | - Zhi Sheng Wang
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China.
| | - Quan Hui Peng
- Institute of Animal Nutrition, Key Laboratory of Bovine Low-Carbon Farming and Safe Production, Sichuan Agricultural University, Ya'an, Sichuan, 625014, PR China.
| |
Collapse
|
1282
|
Bretin A, Lucas C, Larabi A, Dalmasso G, Billard E, Barnich N, Bonnet R, Nguyen HTT. AIEC infection triggers modification of gut microbiota composition in genetically predisposed mice, contributing to intestinal inflammation. Sci Rep 2018; 8:12301. [PMID: 30120269 PMCID: PMC6098085 DOI: 10.1038/s41598-018-30055-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/21/2018] [Indexed: 12/12/2022] Open
Abstract
A high prevalence of adherent-invasive E. coli (AIEC) in the intestinal mucosa of Crohn's disease patients has been shown. AIEC colonize the intestine and induce inflammation in genetically predisposed mouse models including CEABAC10 transgenic (Tg) mice expressing human CEACAM6-receptor for AIEC and eif2ak4-/- mice exhibiting autophagy defect in response to AIEC infection. Here, we aimed at investigating whether gut microbiota modification contributes to AIEC-induced intestinal inflammation in these mouse models. For this, eif2ak4+/+ and eif2ak4-/- mice or CEABAC10 Tg mice invalidated for Eif2ak4 gene (Tg/eif2ak4-/-) or not (Tg/eif2ak4+/+) were infected with the AIEC reference strain LF82 or the non-pathogenic E. coli K12 MG1655 strain. In all mouse groups, LF82 colonized the gut better and longer than MG1655. No difference in fecal microbiota composition was observed in eif2ak4+/+ and eif2ak4-/- mice before infection and at day 1 and 4 post-infection. LF82-infected eif2ak4-/- mice exhibited altered fecal microbiota composition at day 14 and 21 post-infection and increased fecal lipocalin-2 level at day 21 post-infection compared to other groups, indicating that intestinal inflammation developed after microbiota modification. Similar results were obtained for LF82-infected Tg/eif2ak4-/- mice. These results suggest that in genetically predisposed hosts, AIEC colonization might induce chronic intestinal inflammation by altering the gut microbiota composition.
Collapse
Affiliation(s)
- Alexis Bretin
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Cécily Lucas
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Anaïs Larabi
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Guillaume Dalmasso
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Elisabeth Billard
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Nicolas Barnich
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Richard Bonnet
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France
- Centre Hospitalier Universitaire (CHU), Clermont-Ferrand, 63001, France
| | - Hang Thi Thu Nguyen
- M2iSH (Microbes, intestine, inflammation and Susceptibility of the Host), UMR 1071 Inserm, Université Clermont Auvergne, INRA USC 2018, Clermont-Ferrand, 63001, France.
| |
Collapse
|
1283
|
Lyu T, Liu G, Zhang H, Wang L, Zhou S, Dou H, Pang B, Sha W, Zhang H. Changes in feeding habits promoted the differentiation of the composition and function of gut microbiotas between domestic dogs (Canis lupus familiaris) and gray wolves (Canis lupus). AMB Express 2018; 8:123. [PMID: 30073560 PMCID: PMC6072643 DOI: 10.1186/s13568-018-0652-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/26/2018] [Indexed: 02/08/2023] Open
Abstract
Wolves (Canis lupus) and their domesticated and close relatives, dogs (Canis lupus familiaris), have great differences in their diets and living environments. To the best of our knowledge, the fundamental question of how the abundance and function of the gut microbiota of domestic dogs evolved to adapt to the changes in host feeding habits has yet to be addressed. In this study, our comparative analyses of gut metagenomes showed that the abundance of gut microbiota between the two species have some significant differences. Furthermore, a number of taxa observed in higher numbers in domestic dogs are related to carbohydrate metabolism, which may be because that there were more complicated polysaccharides in dogs diets than that in wolves diets. A significant difference in the abundance of genes encoding glycosyltransferase family 34 (GT34), carbohydrate-binding module family 25 (CBM25), and glycoside hydrolase family 13 (GH13) between the gut microbiota metagenomes of domestic dogs and gray wolves also supported this observation. Furthermore, the domestic dog gut microbiota has greater valine, leucine and isoleucine biosynthesis and nitrogen metabolism. This result showed that compared with wolves, the domestic dog diet contains a smaller amount of animal protein, which is consistent with the dietary composition of wolves and dogs. Our results indicate that the function and abundance of gut microbiota of domestic dogs has been adapted to domestication, which is of great significance for the ability of domestic dogs to adapt to changes in food composition.
Collapse
|
1284
|
Hiippala K, Jouhten H, Ronkainen A, Hartikainen A, Kainulainen V, Jalanka J, Satokari R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018; 10:nu10080988. [PMID: 30060606 PMCID: PMC6116138 DOI: 10.3390/nu10080988] [Citation(s) in RCA: 394] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/19/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023] Open
Abstract
The intestinal microbiota, composed of pro- and anti-inflammatory microbes, has an essential role in maintaining gut homeostasis and functionality. An overly hygienic lifestyle, consumption of processed and fiber-poor foods, or antibiotics are major factors modulating the microbiota and possibly leading to longstanding dysbiosis. Dysbiotic microbiota is characterized to have altered composition, reduced diversity and stability, as well as increased levels of lipopolysaccharide-containing, proinflammatory bacteria. Specific commensal species as novel probiotics, so-called next-generation probiotics, could restore the intestinal health by means of attenuating inflammation and strengthening the epithelial barrier. In this review we summarize the latest findings considering the beneficial effects of the promising commensals across all major intestinal phyla. These include the already well-known bifidobacteria, which use extracellular structures or secreted substances to promote intestinal health. Faecalibacterium prausnitzii, Roseburia intestinalis, and Eubacterium hallii metabolize dietary fibers as major short-chain fatty acid producers providing energy sources for enterocytes and achieving anti-inflammatory effects in the gut. Akkermansia muciniphila exerts beneficial action in metabolic diseases and fortifies the barrier function. The health-promoting effects of Bacteroides species are relatively recently discovered with the findings of excreted immunomodulatory molecules. These promising, unconventional probiotics could be a part of biotherapeutic strategies in the future.
Collapse
Affiliation(s)
- Kaisa Hiippala
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Hanne Jouhten
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Aki Ronkainen
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Anna Hartikainen
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Veera Kainulainen
- Pharmacology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Jonna Jalanka
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Reetta Satokari
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
1285
|
Yu LCH, Wei SC, Ni YH. Impact of microbiota in colorectal carcinogenesis: lessons from experimental models. Intest Res 2018; 16:346-357. [PMID: 30090033 PMCID: PMC6077307 DOI: 10.5217/ir.2018.16.3.346] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023] Open
Abstract
A role of gut microbiota in colorectal cancer (CRC) growth was first suggested in germ-free rats almost 50 years ago, and the existence of disease-associated bacteria (termed pathobionts) had becoming increasingly evident from experimental data of fecal transplantation, and microbial gavage or monoassociation. Altered bacterial compositions in fecal and mucosal specimens were observed in CRC patients compared to healthy subjects. Microbial fluctuations were found at various cancer stages; an increase of bacterial diversity was noted in the adenoma specimens, while a reduction of bacterial richness was documented in CRC samples. The bacterial species enriched in the human cancerous tissues included Escherichia coli, Fusobacterium nucleatum, and enterotoxigenic Bacteroides fragilis. The causal relationship of gut bacteria in tumorigenesis was established by introducing particular bacterial strains in in situ mouse CRC models. Detailed experimental protocols of bacterial gavage and the advantages and caveats of different experimental models are summarized in this review. The microbial genotoxins, enterotoxins, and virulence factors implicated in the mechanisms of bacteria-driven tumorigenesis are described. In conclusion, intestinal microbiota is involved in colon tumorigenesis. Bacteria-targeting intervention would be the next challenge for CRC.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shu-Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hsuan Ni
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
1286
|
Geerlings SY, Kostopoulos I, de Vos WM, Belzer C. Akkermansia muciniphila in the Human Gastrointestinal Tract: When, Where, and How? Microorganisms 2018; 6:microorganisms6030075. [PMID: 30041463 PMCID: PMC6163243 DOI: 10.3390/microorganisms6030075] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/03/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023] Open
Abstract
Akkermansia muciniphila is a mucin-degrading bacterium of the phylum Verrucomicrobia. Its abundance in the human intestinal tract is inversely correlated to several disease states. A. muciniphila resides in the mucus layer of the large intestine, where it is involved in maintaining intestinal integrity. We explore the presence of Akkermansia-like spp. based on its 16S rRNA sequence and metagenomic signatures in the human body so as to understand its colonization pattern in time and space. A. muciniphila signatures were detected in colonic samples as early as a few weeks after birth and likely could be maintained throughout life. The sites where Akkermansia-like sequences (including Verrucomicrobia phylum and/or Akkermansia spp. sequences found in the literature) were detected apart from the colon included human milk, the oral cavity, the pancreas, the biliary system, the small intestine, and the appendix. The function of Akkermansia-like spp. in these sites may differ from that in the mucosal layer of the colon. A. muciniphila present in the appendix or in human milk could play a role in the re-colonization of the colon or breast-fed infants, respectively. In conclusion, even though A. muciniphila is most abundantly present in the colon, the presence of Akkermansia-like spp. along the digestive tract indicates that this bacterium might have more functions than those currently known.
Collapse
Affiliation(s)
- Sharon Y Geerlings
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708WE Wageningen, The Netherlands.
| | - Ioannis Kostopoulos
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708WE Wageningen, The Netherlands.
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708WE Wageningen, The Netherlands.
- Immunobiology Research Program, Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, 00014 Helsinki, Finland.
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708WE Wageningen, The Netherlands.
| |
Collapse
|
1287
|
Salem I, Ramser A, Isham N, Ghannoum MA. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front Microbiol 2018; 9:1459. [PMID: 30042740 PMCID: PMC6048199 DOI: 10.3389/fmicb.2018.01459] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The adult intestine hosts a myriad of diverse bacterial species that reside mostly in the lower gut maintaining a symbiosis with the human habitat. In the current review, we describe the neoteric advancement in our comprehension of how the gut microbiota communicates with the skin as one of the main regulators in the gut-skin axis. We attempted to explore how this potential link affects skin differentiation and keratinization, its influence on modulating the cutaneous immune response in various diseases, and finally how to take advantage of this communication in the control of different skin conditions.
Collapse
Affiliation(s)
- Iman Salem
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Amy Ramser
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Nancy Isham
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Mahmoud A. Ghannoum
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
1288
|
Park SH, Perrotta A, Hanning I, Diaz-Sanchez S, Pendleton S, Alm E, Ricke SC. Pasture flock chicken cecal microbiome responses to prebiotics and plum fiber feed amendments. Poult Sci 2018; 96:1820-1830. [PMID: 28339946 DOI: 10.3382/ps/pew441] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 11/16/2016] [Indexed: 12/22/2022] Open
Abstract
When prebiotics and other fermentation substrates are delivered to animals as feed supplements, the typical goal is to improve weight gain and feed conversion. In this work, we examined pasture flock chicken cecal contents using next generation sequencing (NGS) to identify and understand the composition of the microbiome when prebiotics and fermentation substrates were supplemented. We generated 16S rRNA sequencing data for 120 separate cecal samples from groups of chickens receiving one of 3 prebiotics or fiber feed additives. The data indicated that respective feed additives enrich for specific bacterial community members and modulate the diversity of the microbiome. We applied synthetic learning in microbial ecology (SLiME) analysis to interpret 16S rRNA microbial community data and identify specific bacterial operational taxonomic units (OTU) that are predictive of the particular feed additives used in these experiments. The results suggest that feed can influence microbiome composition in a predictable way, and thus diet may have indirect effects on weight gain and feed conversion through the microbiome.
Collapse
Affiliation(s)
- S H Park
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville.,Center for Food Safety and Department of Food Science, University of Arkansas, Fayetteville
| | - A Perrotta
- Department of Civil and Environmental Engineering and Biological Engineering, and the Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA
| | - I Hanning
- Department of Science, Lincoln International Academy, Managua, Nicaragua.,The Graduate School of Genome Sciences and Technology, University of Tennessee, Knoxville
| | - S Diaz-Sanchez
- Department of Food Science and Technology, University of Tennessee, Knoxville.,SaBio IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - S Pendleton
- Department of Food Science and Technology, University of Tennessee, Knoxville
| | - E Alm
- Department of Civil and Environmental Engineering and Biological Engineering, and the Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA
| | - S C Ricke
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville.,Center for Food Safety and Department of Food Science, University of Arkansas, Fayetteville
| |
Collapse
|
1289
|
Bang SJ, Kim G, Lim MY, Song EJ, Jung DH, Kum JS, Nam YD, Park CS, Seo DH. The influence of in vitro pectin fermentation on the human fecal microbiome. AMB Express 2018; 8:98. [PMID: 29909506 PMCID: PMC6004267 DOI: 10.1186/s13568-018-0629-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 12/21/2022] Open
Abstract
Pectin is a complex dietary fiber and a prebiotic. To investigate pectin-induced changes in the gut microbiome and their effects on the short chain fatty acids (SCFAs) production, we performed in vitro pectin fermentation using the feces of three Korean donors. The pectin degradations in all three donors were observed. While the donors displayed differences in baseline gut microbiota composition, commonly increased bacteria after pectin fermentation included Lachnospira, Dorea, Clostridium, and Sutterella. Regarding SCFAs, acetate levels rapidly increased with incubation with pectin, and butyrate levels also increased after 6 h of incubation. The results suggest that pectin fermentation increases bacterial species belonging to Clostridium cluster XIV (Lachnospira, Dorea, and Clostridium), with Lachnospira displaying the greatest increase. The results also confirm that pectin fermentation leads to the production of acetate and butyrate.
Collapse
|
1290
|
Rahmouni O, Vignal C, Titécat M, Foligné B, Pariente B, Dubuquoy L, Desreumaux P, Neut C. High carriage of adherent invasive E. coli in wildlife and healthy individuals. Gut Pathog 2018; 10:23. [PMID: 29946365 PMCID: PMC6001069 DOI: 10.1186/s13099-018-0248-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/23/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Adherent invasive Escherichia coli (AIEC) are suspected to be involved in the pathogenesis of inflammatory bowel diseases. Since AIEC was first described in 1999, despite important progress on its genomic and immune characterizations, some crucial questions remain unanswered, such as whether there exists a natural reservoir, or whether there is asymptomatic carriage. The ECOR collection, including E. coli strains isolated mainly from the gut of healthy humans and animals, constitutes an ideal tool to investigate AIEC prevalence in healthy condition. A total of 61 E. coli strains were examined for characteristics of AIEC. METHODS The adhesion, invasion and intramacrophage replication capabilities (AIEC phenotype) of 61 intestinal E. coli strains were determined. The absence of virulence-associated diarrheagenic E. coli pathotypes (EPEC, ETEC, EIEC, EHEC, DAEC, EAEC), and uropathogenic E. coli was checked. RESULTS Out of 61 intestinal strains, 13 (21%) exhibit the AIEC phenotype, 7 are from human origin and 6 are from animal origin. Prevalence of AIEC strains is about 24 and 19% in healthy humans and animals respectively. These strains are highly genetically diverse as they are distributed among the main described phylogroups. Among E. coli strains from the ECOR collection, we also detected strains able to detach I-407 cells. CONCLUSIONS Our study described for the first time AIEC strains isolated from the feces of healthy humans and animals.
Collapse
Affiliation(s)
- Oumaïra Rahmouni
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
| | - Cécile Vignal
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
| | - Marie Titécat
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
- Centre de Biologie Pathologie Génétique, CHU Lille, Lille, France
| | - Benoît Foligné
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
| | - Benjamin Pariente
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
- Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Claude Huriez, CHU Lille, 59037 Lille, France
| | - Laurent Dubuquoy
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
| | - Pierre Desreumaux
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
- Service des Maladies de l’Appareil Digestif et de la Nutrition, Hôpital Claude Huriez, CHU Lille, 59037 Lille, France
| | - Christel Neut
- Lille Inflammation Research International Center, UMR 995 Inserm, Lille University, CHRU Lille, Lille, France
- Laboratoire de Bactériologie, 3, Rue de Pr. Laguesse, B.P. 83, 59006 Lille Cedex, France
| |
Collapse
|
1291
|
Xu J, Chen N, Wu Z, Song Y, Zhang Y, Wu N, Zhang F, Ren X, Liu Y. 5-Aminosalicylic Acid Alters the Gut Bacterial Microbiota in Patients With Ulcerative Colitis. Front Microbiol 2018; 9:1274. [PMID: 29951050 PMCID: PMC6008376 DOI: 10.3389/fmicb.2018.01274] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/24/2018] [Indexed: 12/15/2022] Open
Abstract
Background: The aim of this study was to clarify the effect of 5-aminosalicylic acid (5-ASA) treatment on gut bacterial microbiota in patients with ulcerative colitis (UC). Methods: A total of 57 UC patients, including 20 untreated and 37 5-ASA-treated, were recruited into an exploration cohort. We endoscopically collected both non-inflamed and inflamed mucosal samples from all patients, and compared the gut bacterial profiles using 16S rDNA sequencing. Ten untreated UC patients were then treated with 5-ASA and subsequently recruited for an independent validation study to confirm the acquired data. Results: In untreated UC patients, compared with those in non-inflamed mucosae, Firmicutes (such as Enterococcus) were decreased and Proteobacteria (e.g., Escherichia–Shigella) were increased in the inflamed mucosae. Compared with the inflamed mucosae of untreated UC patients, there was a higher abundance of Firmicutes (e.g., Enterococcus) and lower Proteobacteria (Escherichia–Shigella) in the inflamed mucosae of 5-ASA treated UC patients. In the validation cohort, after administration of 5-ASA, bacterial alteration was consistent with these data. Furthermore, there was a skewed negative correlation between Escherichia–Shigella and bacterial genera of Firmicutes in the inflamed mucosae. 5-ASA treatment decreased the strength of bacterial correlation and weakened the skewed negative correlation pattern. Conclusion: The microbial dysbiosis (mainly characterized by an increased abundance in the Escherichia–Shigella genus) and the skewed negative correlation between Escherichia–Shigella and bacterial genera of Firmicutes are two characteristics of the inflamed mucosae of UC patients. 5-ASA treatment decreases Escherichia–Shigella and weakens the skewed correlations, which may be related to its treatment efficiency.
Collapse
Affiliation(s)
- Jun Xu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Zhe Wu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Yang Song
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Yifan Zhang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Na Wu
- Institute of Clinical Molecular Biology and Central Laboratory, Peking University People's Hospital, Beijing, China
| | - Feng Zhang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Xinhua Ren
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| |
Collapse
|
1292
|
Escherichia coli B2 strains prevalent in inflammatory bowel disease patients have distinct metabolic capabilities that enable colonization of intestinal mucosa. BMC SYSTEMS BIOLOGY 2018; 12:66. [PMID: 29890970 PMCID: PMC5996543 DOI: 10.1186/s12918-018-0587-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/21/2018] [Indexed: 01/04/2023]
Abstract
Background Escherichia coli is considered a leading bacterial trigger of inflammatory bowel disease (IBD). E. coli isolates from IBD patients primarily belong to phylogroup B2. Previous studies have focused on broad comparative genomic analysis of E. coli B2 isolates, and identified virulence factors that allow B2 strains to reside within human intestinal mucosa. Metabolic capabilities of E. coli strains have been shown to be related to their colonization site, but remain unexplored in IBD-associated strains. Results In this study, we utilized pan-genome analysis and genome-scale models (GEMs) of metabolism to study metabolic capabilities of IBD-associated E. coli B2 strains. The study yielded three results: i) Pan-genome analysis of 110 E. coli strains (including 53 isolates from IBD studies) revealed discriminating metabolic genes between B2 strains and other strains; ii) Both comparative genomic analysis and GEMs suggested that B2 strains have an advantage in degrading and utilizing sugars derived from mucus glycan, and iii) GEMs revealed distinct metabolic features in B2 strains that potentially allow them to utilize energy more efficiently. For example, B2 strains lack the enzymes to degrade amadori products, but instead rely on neighboring bacteria to convert these substrates into a more readily usable and potentially less sought after product. Conclusions Taken together, these results suggest that the metabolic capabilities of B2 strains vary significantly from those of other strains, enabling B2 strains to colonize intestinal mucosa.The results from this study motivate a broad experimental assessment of the nutritional effects on E. coli B2 pathophysiology in IBD patients. Electronic supplementary material The online version of this article (10.1186/s12918-018-0587-5) contains supplementary material, which is available to authorized users.
Collapse
|
1293
|
Chia LW, Hornung BVH, Aalvink S, Schaap PJ, de Vos WM, Knol J, Belzer C. Deciphering the trophic interaction between Akkermansia muciniphila and the butyrogenic gut commensal Anaerostipes caccae using a metatranscriptomic approach. Antonie Van Leeuwenhoek 2018; 111:859-873. [PMID: 29460206 PMCID: PMC5945754 DOI: 10.1007/s10482-018-1040-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 02/02/2018] [Indexed: 12/26/2022]
Abstract
Host glycans are paramount in regulating the symbiotic relationship between humans and their gut bacteria. The constant flux of host-secreted mucin at the mucosal layer creates a steady niche for bacterial colonization. Mucin degradation by keystone species subsequently shapes the microbial community. This study investigated the transcriptional response during mucin-driven trophic interaction between the specialised mucin-degrader Akkermansia muciniphila and a butyrogenic gut commensal Anaerostipes caccae. A. muciniphila monocultures and co-cultures with non-mucolytic A. caccae from the Lachnospiraceae family were grown anaerobically in minimal media supplemented with mucin. We analysed for growth, metabolites (HPLC analysis), microbial composition (quantitative reverse transcription PCR), and transcriptional response (RNA-seq). Mucin degradation by A. muciniphila supported the growth of A. caccae and concomitant butyrate production predominantly via the acetyl-CoA pathway. Differential expression analysis (DESeq 2) showed the presence of A. caccae induced changes in the A. muciniphila transcriptional response with increased expression of mucin degradation genes and reduced expression of ribosomal genes. Two putative operons that encode for uncharacterised proteins and an efflux system, and several two-component systems were also differentially regulated. This indicated A. muciniphila changed its transcriptional regulation in response to A. caccae. This study provides insight to understand the mucin-driven microbial ecology using metatranscriptomics. Our findings show that the expression of mucolytic enzymes by A. muciniphila increases upon the presence of a community member. This could indicate its role as a keystone species that supports the microbial community in the mucosal environment by increasing the availability of mucin sugars.
Collapse
Affiliation(s)
- Loo Wee Chia
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Bastian V H Hornung
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Steven Aalvink
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Peter J Schaap
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- RPU Immunobiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, 00290, Helsinki, Finland
| | - Jan Knol
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
- Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| |
Collapse
|
1294
|
Zengler K, Zaramela LS. The social network of microorganisms - how auxotrophies shape complex communities. Nat Rev Microbiol 2018; 16:383-390. [PMID: 29599459 PMCID: PMC6059367 DOI: 10.1038/s41579-018-0004-5] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microorganisms engage in complex interactions with other organisms and their environment. Recent studies have shown that these interactions are not limited to the exchange of electron donors. Most microorganisms are auxotrophs, thus relying on external nutrients for growth, including the exchange of amino acids and vitamins. Currently, we lack a deeper understanding of auxotrophies in microorganisms and how nutrient requirements differ between different strains and different environments. In this Opinion article, we describe how the study of auxotrophies and nutrient requirements among members of complex communities will enable new insights into community composition and assembly. Understanding this complex network over space and time is crucial for developing strategies to interrogate and shape microbial communities.
Collapse
Affiliation(s)
- Karsten Zengler
- Department of Pediatrics, Division of Host-Microbe Systems & Therapeutics, University of California, San Diego, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, CA, USA.
| | - Livia S Zaramela
- Department of Pediatrics, Division of Host-Microbe Systems & Therapeutics, University of California, San Diego, CA, USA
| |
Collapse
|
1295
|
Migliore F, Macchi R, Landini P, Paroni M. Phagocytosis and Epithelial Cell Invasion by Crohn's Disease-Associated Adherent-Invasive Escherichia coli Are Inhibited by the Anti-inflammatory Drug 6-Mercaptopurine. Front Microbiol 2018; 9:964. [PMID: 29867868 PMCID: PMC5961443 DOI: 10.3389/fmicb.2018.00964] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/24/2018] [Indexed: 12/22/2022] Open
Abstract
Adherent-invasive Escherichia coli (AIEC) strains are overrepresented in the dysbiotic microbiota of Crohn’s disease (CD) patients, and contribute to the onset of the chronic inflammation typical of the disease. However, the effects of anti-inflammatory drugs used for CD treatment on AIEC virulence have not yet been investigated. In this report, we show that exposure of AIEC LF82 strain to amino-6-mercaptopurine (6-MP) riboside, one of the most widely used anti-inflammatory drugs in CD, impairs its ability to adhere to, and consequently to invade, human epithelial cells. Notably, phagocytosis of LF82 treated with 6-MP by human macrophages is also reduced, suggesting that 6-MP affects AIEC cell surface determinants involved both in interaction with epithelial cells and in uptake by macrophages. Since a main target of 6-MP in bacterial cells is the inhibition of the important signal molecule c-di-GMP, we also tested whether perturbations in cAMP, another major signaling pathway in E. coli, might have similar effects on interactions with human cells. To this aim, we grew LF82 in the presence of glucose, which leads to inhibition of cAMP synthesis. Growth in glucose-supplemented medium resulted in a reduction in AIEC adhesion to epithelial cells and uptake by macrophages. Consistent with these results, both 6-MP and glucose can affect expression of cell adhesion-related genes, such as the csg genes, encoding thin aggregative fimbriae (curli). In addition, glucose strongly inhibits expression of the fim operon, encoding type 1 pili, a known AIEC determinant for adhesion to human cells. To further investigate whether 6-MP can indeed inhibit c-di-GMP signaling in AIEC, we performed biofilm and motility assays and determination of extracellular polysaccharides. 6-MP clearly affected biofilm formation and cellulose production, but also, unexpectedly, reduced cell motility, itself an important virulence factor for AIEC. Our results provide strong evidence that 6-MP can affect AIEC-host cell interaction by acting on the bacterial cell, thus strengthening the hypothesis that mercaptopurines might promote CD remission also by affecting gut microbiota composition and/or physiology, and suggesting that novel drugs targeting bacterial virulence and signaling might be effective in preventing chronic inflammation in CD.
Collapse
Affiliation(s)
- Federica Migliore
- Dipartimento di Bioscienze, Università degli studi di Milano, Milan, Italy
| | - Raffaella Macchi
- Dipartimento di Bioscienze, Università degli studi di Milano, Milan, Italy
| | - Paolo Landini
- Dipartimento di Bioscienze, Università degli studi di Milano, Milan, Italy
| | - Moira Paroni
- Dipartimento di Bioscienze, Università degli studi di Milano, Milan, Italy
| |
Collapse
|
1296
|
Lagier JC, Dubourg G, Million M, Cadoret F, Bilen M, Fenollar F, Levasseur A, Rolain JM, Fournier PE, Raoult D. Culturing the human microbiota and culturomics. Nat Rev Microbiol 2018; 16:540-550. [PMID: 29937540 DOI: 10.1038/s41579-018-0041-0] [Citation(s) in RCA: 519] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gut microbiota has an important role in the maintenance of human health and in disease pathogenesis. This importance was realized through the advent of omics technologies and their application to improve our knowledge of the gut microbial ecosystem. In particular, the use of metagenomics has revealed the diversity of the gut microbiota, but it has also highlighted that the majority of bacteria in the gut remain uncultured. Culturomics was developed to culture and identify unknown bacteria that inhabit the human gut as a part of the rebirth of culture techniques in microbiology. Consisting of multiple culture conditions combined with the rapid identification of bacteria, the culturomic approach has enabled the culture of hundreds of new microorganisms that are associated with humans, providing exciting new perspectives on host-bacteria relationships. In this Review, we discuss why and how culturomics was developed. We describe how culturomics has extended our understanding of bacterial diversity and then explore how culturomics can be applied to the study of the human microbiota and the potential implications for human health.
Collapse
Affiliation(s)
- Jean-Christophe Lagier
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Grégory Dubourg
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Matthieu Million
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Frédéric Cadoret
- Assistance Publique-Hôpitaux de Marseille, IHU Méditerranée Infection, Marseille, France
| | - Melhem Bilen
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France.,Fondation Méditerranée Infection, IHU Méditerranée Infection, Marseille, France
| | - Florence Fenollar
- Aix Marseille Université, IRD, AP-HM, VITROME, IHU Méditerranée Infection, Marseille, France
| | - Anthony Levasseur
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Jean-Marc Rolain
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Pierre-Edouard Fournier
- Aix Marseille Université, IRD, AP-HM, VITROME, IHU Méditerranée Infection, Marseille, France
| | - Didier Raoult
- Aix Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée Infection, Marseille, France.
| |
Collapse
|
1297
|
Guo YF, Shu L, Tan ZJ. Role of intestinal Clostridium in pathogenesis and treatment of diarrhea. Shijie Huaren Xiaohua Zazhi 2018; 26:693-699. [DOI: 10.11569/wcjd.v26.i12.693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Yan-Fang Guo
- the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan Province, China
| | - Lan Shu
- the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan Province, China
| | - Zhou-Jin Tan
- Department of Microbiology, Hu'nan University of Chinese Medicine, Changsha 410208, Hunan Province, China
| |
Collapse
|
1298
|
Wang SL, Shao BZ, Zhao SB, Fang J, Gu L, Miao CY, Li ZS, Bai Y. Impact of Paneth Cell Autophagy on Inflammatory Bowel Disease. Front Immunol 2018; 9:693. [PMID: 29675025 PMCID: PMC5895641 DOI: 10.3389/fimmu.2018.00693] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/21/2018] [Indexed: 12/19/2022] Open
Abstract
Intestinal mucosal barrier, mainly consisting of the mucus layer and epithelium, functions in absorbing nutrition as well as prevention of the invasion of pathogenic microorganisms. Paneth cell, an important component of mucosal barrier, plays a vital role in maintaining the intestinal homeostasis by producing antimicrobial materials and controlling the host-commensal balance. Current evidence shows that the dysfunction of intestinal mucosal barrier, especially Paneth cell, participates in the onset and progression of inflammatory bowel disease (IBD). Autophagy, a cellular stress response, involves various physiological processes, such as secretion of proteins, production of antimicrobial peptides, and degradation of aberrant organelles or proteins. In the recent years, the roles of autophagy in the pathogenesis of IBD have been increasingly studied. Here in this review, we mainly focus on describing the roles of Paneth cell autophagy in IBD as well as several popular autophagy-related genetic variants in Penath cell and the related therapeutic strategies against IBD.
Collapse
Affiliation(s)
- Shu-Ling Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Bo-Zong Shao
- Department of Pharmocology, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Sheng-Bing Zhao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Jun Fang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lun Gu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmocology, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University and Naval Medical University, Shanghai, China
| |
Collapse
|
1299
|
Bazin T, Hooks KB, Barnetche T, Truchetet ME, Enaud R, Richez C, Dougados M, Hubert C, Barré A, Nikolski M, Schaeverbeke T. Microbiota Composition May Predict Anti-Tnf Alpha Response in Spondyloarthritis Patients: an Exploratory Study. Sci Rep 2018; 8:5446. [PMID: 29615661 PMCID: PMC5882885 DOI: 10.1038/s41598-018-23571-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023] Open
Abstract
Spondyloarthritis (SpA) pathophysiology remains largely unknown. While the association with genetic factors has been established for decades, the influence of gut microbiota is only an emerging direction of research. Despite the remarkable efficacy of anti-TNF-α treatments, non-responders are frequent and no predictive factors of patient outcome have been identified. Our objective was to investigate the modifications of intestinal microbiota composition in patients suffering from SpA three months after an anti-TNF-α treatment. We performed 16S rDNA sequencing of 38 stool samples from 19 spondyloarthritis patients before and three months after anti-TNF-α treatment onset. SpA activity was assessed at each time using ASDAS and BASDAI scores. Some modifications of the microbiota composition were observed after three months of anti-TNF-α treatment, but no specific taxon was modified, whatever the clinical response. We identified a particular taxonomic node before anti-TNF-α treatment that can predict the clinical response as a biomarker, with a higher proportion of Burkholderiales order in future responder patients. This study suggests a cross-influence between anti-TNF-α treatment and intestinal microbiota. If its results are confirmed on larger groups of patients, it may pave the way to the development of predictive tests suitable for clinical practices.
Collapse
Affiliation(s)
- Thomas Bazin
- Univ. Bordeaux, INRA, Mycoplasmal and chlamydial infections in humans, EA 3671, 33000, Bordeaux, France
- Bordeaux Hospital University Center, Department of Hepato-gastroenterology, 33600, Pessac, France
| | - Katarzyna B Hooks
- Univ. Bordeaux, Bordeaux Bioinformatics Center, 33000, Bordeaux, France
- Univ. Bordeaux, CNRS, Immunoconcept, UMR 5164, 33000, Bordeaux, France
| | - Thomas Barnetche
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France
| | - Marie-Elise Truchetet
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France
| | - Raphaël Enaud
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, FHU ACRONIM, Laboratoire 8 de Parasitologie-Mycologie, F-33000, Bordeaux, France
- CHU Bordeaux, Unité d'Hépatologie, Gastroentérologie et Nutrition Pédiatriques, CRCM Pédiatrique, Service 10 de Rhumatologie, Service d'Exploration Fonctionnelle Respiratoire, 33000, Bordeaux, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, F-33000, Bordeaux, France
| | - Christophe Richez
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France
| | - Maxime Dougados
- AP-HP, Cochin Hospital University Center, Department of Rheumatology, 75014, Paris, France
| | - Christophe Hubert
- Univ. Bordeaux, INSERM, Rare Diseases, genetic and metabolism, U1211, 33000, Bordeaux, France
- Univ. Bordeaux, Genome Transcriptome Facility of Bordeaux, 33000, Bordeaux, France
| | - Aurélien Barré
- Univ. Bordeaux, Bordeaux Bioinformatics Center, 33000, Bordeaux, France
| | - Macha Nikolski
- Univ. Bordeaux, Bordeaux Bioinformatics Center, 33000, Bordeaux, France
- Univ. Bordeaux, CNRS, LaBRI, UMR 5800, 33400, Talence, France
| | - Thierry Schaeverbeke
- Univ. Bordeaux, INRA, Mycoplasmal and chlamydial infections in humans, EA 3671, 33000, Bordeaux, France.
- Bordeaux Hospital University Center, Department of Rheumatology, 33000, Bordeaux, France.
| |
Collapse
|
1300
|
Testa A, Rispo A, Imperatore N, Nardone OM, Trinchese G, Cavaliere G, Castiglione F, Mollica MP. Gut microbiota and Crohn’s disease. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2018. [DOI: 10.3233/mnm-17182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION: Crohn’s disease (CD) is characterized by a chronic inflammation of the gastrointestinal tract causing abdominal pain, diarrhea, weight loss and systemic symptoms. Although the etiology of this disease is unknown, current knowledge suggests a multifactorial genesis involving genetic, environmental and immunological factors. EVIDENCE ACQUISITION: We focused our attention on critical analysis of the recent literature on the role of gut microbiota in inflammatory bowel disease (IBD), by evaluating the differences of composition, functions and role of intestinal flora. In particular, we focused on evidences about the interaction between gut microbiota and pathogenesis of IBD. In this setting, we conducted a PUBMED search for guidelines, systematic reviews (SR) and primary studies. EVIDENCE SYNTHESIS: Some data suggest that, in a significant percentage of patients, the microbiota plays an important role in the genesis and maintenance of CD. Probiotic supplementation and antibiotic treatment appear to be a valid therapeutic approach, although the clinical data remain controversial. CONCLUSIONS: Despite the exciting and growing research on the role of gut microbiota in IBD, our knowledge remains fairly limited. Further studies are needed to measure the diversity, function and resistance to antibiotics of the intestinal microbiota in CD.
Collapse
Affiliation(s)
- Anna Testa
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Antonio Rispo
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Nicola Imperatore
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Olga Maria Nardone
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Giovanna Trinchese
- Department of Biology, University of Naples “Federico II”, Naples, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples “Federico II”, Naples, Italy
| | - Fabiana Castiglione
- Department of Clinical Medicine and Surgery, Gastroenterology, School of Medicine “Federico II” of Naples, Naples, Italy
| | - Maria Pina Mollica
- Department of Biology, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|