1
|
Zangara MT, Darwish L, Coombes BK. Characterizing the Pathogenic Potential of Crohn's Disease-Associated Adherent-Invasive Escherichia coli. EcoSal Plus 2023; 11:eesp00182022. [PMID: 37220071 PMCID: PMC10729932 DOI: 10.1128/ecosalplus.esp-0018-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/04/2023] [Indexed: 01/28/2024]
Abstract
The microbiome of Crohn's disease (CD) patients is composed of a microbial community that is considered dysbiotic and proinflammatory in nature. The overrepresentation of Enterobacteriaceae species is a common feature of the CD microbiome, and much attention has been given to understanding the pathogenic role this feature plays in disease activity. Over 2 decades ago, a new Escherichia coli subtype called adherent-invasive E. coli (AIEC) was isolated and linked to ileal Crohn's disease. Since the isolation of the first AIEC strain, additional AIEC strains have been isolated from both inflammatory bowel disease (IBD) patients and non-IBD individuals using the original in vitro phenotypic characterization methods. Identification of a definitive molecular marker of the AIEC pathotype has been elusive; however, significant advancements have been made in understanding the genetic, metabolic, and virulence determinants of AIEC infection biology. Here, we review the current knowledge of AIEC pathogenesis to provide additional, objective measures that could be considered in defining AIEC and their pathogenic potential.
Collapse
Affiliation(s)
- Megan T. Zangara
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Lena Darwish
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Brian K. Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| |
Collapse
|
2
|
Guillaume D, Racha B, Sandrine B, Etienne R, Laurent G, Virginie B, Pierre SS, Amine G, Vincent G, Nicolas B, Julien D, Richard B. Genes mcr improve the intestinal fitness of pathogenic E. coli and balance their lifestyle to commensalism. MICROBIOME 2023; 11:12. [PMID: 36670449 PMCID: PMC9863213 DOI: 10.1186/s40168-022-01457-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 12/22/2022] [Indexed: 05/10/2023]
Abstract
BACKGROUND The plasmid-mediated resistance gene mcr-1 confers colistin resistance in Escherichia coli and paves the way for the evolution to pan-drug resistance. We investigated the impact of mcr-1 in gut colonization in the absence of antibiotics using isogenic E. coli strains transformed with a plasmid encoding or devoid of mcr-1. RESULTS In gnotobiotic and conventional mice, mcr-1 significantly enhanced intestinal anchoring of E. coli but impaired their lethal effect. This improvement of intestinal fitness was associated with a downregulation of intestinal inflammatory markers and the preservation of intestinal microbiota composition. The mcr-1 gene mediated a cross-resistance to antimicrobial peptides secreted by the microbiota and intestinal epithelial cells (IECs), enhanced E. coli adhesion to IECs, and decreased the proinflammatory activity of both E. coli and its lipopolysaccharides. CONCLUSION Overall, mcr-1 changed multiple facets of bacterial behaviour and appeared as a factor enhancing commensal lifestyle and persistence in the gut even in the absence of antibiotics. Video Abstract.
Collapse
Affiliation(s)
- Dalmasso Guillaume
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Beyrouthy Racha
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
- Centre de référence de la résistance aux antibiotiques, Centre Hospitalier Universitaire, 58 place Montalembert, 63000 Clermont-Ferrand, France
| | - Brugiroux Sandrine
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Ruppé Etienne
- Université de Paris, IAME, INSERM, F-75018 Paris, France
- AP-HP, Hôpital Bichat, DEBRC, F-75018 Paris, France
| | - Guillouard Laurent
- Centre de référence de la résistance aux antibiotiques, Centre Hospitalier Universitaire, 58 place Montalembert, 63000 Clermont-Ferrand, France
| | - Bonnin Virginie
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Saint-Sardos Pierre
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Ghozlane Amine
- Hub de Bioinformatique et Biostatistique—Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Gaumet Vincent
- IMOST, UMR 1240 Inserm, Université Clermont Auvergne, 58 Rue Montalembert, 63005 Clermont-Ferrand, France
| | - Barnich Nicolas
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Delmas Julien
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Bonnet Richard
- Université Clermont Auvergne, Inserm U1071, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l’Hôte (M2iSH), Centre de Recherche en Nutrition Humaine Auvergne, 28 place Henri Dunant, 63001 Clermont-Ferrand, France
- Centre de référence de la résistance aux antibiotiques, Centre Hospitalier Universitaire, 58 place Montalembert, 63000 Clermont-Ferrand, France
| |
Collapse
|
3
|
Sun F, Sun Y, Wang Y, Yuan Q, Xiong L, Feng W, Xia P. Role of Penicillin-Binding Protein 1b in the Biofilm Inhibitory Efficacy of Ceftazidime Against Escherichia coli. Curr Microbiol 2022; 79:271. [PMID: 35881255 DOI: 10.1007/s00284-022-02966-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 07/05/2022] [Indexed: 11/24/2022]
Abstract
Penicillin-binding proteins (PBPs) play an important role in bacterial biofilm formation and are the targets of β-lactam antibiotics. This study aimed to investigate the effect of the β-lactam antibiotic ceftazidime (CAZ) at subminimal inhibitory concentration (sub-MIC) on the biofilm formation of Escherichia coli by targeting PBPs. In this study, PBP1a (encoded by mrcA), PBP1b (encoded by mrcB) and PBP3 (encoded by ftsI), which have high affinity for CAZ, were deleted from the E. coli strain. The mrcB mutant showed lower adhesion, biofilm formation and swimming motility, whereas the knockout of mrcA or ftsI had no obvious influence on the biofilm-associated indicators mentioned above. After treatment with sub-MIC of CAZ, the adhesion, biofilm formation and swimming motility of the mrcB-mutant strain were not different or were slightly reduced compared with those of the untreated group. However, sub-MIC of CAZ still significantly inhibited these biofilm-associated indicators in mrcA- and ftsI-mutant strains. In addition, consistent with the bacterial motility results, the deletion of the mrcB gene reduced the flagellar numbers and the expression of flagellar structural genes, but flagellum-related indicators in the mrcB-mutant strain treated with CAZ were similar to those in the untreated group. Bioinformatic analysis showed that CAZ binds to Lys287, Lys274, Glu281, and Arg286 in PBP1b. Taken together, these results suggest that CAZ reduced flagellar synthesis and bacterial motility by binding with PBP1b and thereby inhibited the adhesion and biofilm formation of E. coli.
Collapse
Affiliation(s)
- Fengjun Sun
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30, Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yixuan Sun
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30, Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Department of Pharmacy, Chongqing Municipal People's Hospital, Chongqing, 400014, China
| | - Yu Wang
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30, Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Qian Yuan
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30, Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Lirong Xiong
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30, Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Wei Feng
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30, Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
| | - Peiyuan Xia
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30, Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
| |
Collapse
|
4
|
Checkpoints That Regulate Balanced Biosynthesis of Lipopolysaccharide and Its Essentiality in Escherichia coli. Int J Mol Sci 2021; 23:ijms23010189. [PMID: 35008618 PMCID: PMC8745692 DOI: 10.3390/ijms23010189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022] Open
Abstract
The outer membrane (OM) of Gram-negative bacteria, such as Escherichia coli, is essential for their viability. Lipopolysaccharide (LPS) constitutes the major component of OM, providing the permeability barrier, and a tight balance exists between LPS and phospholipids amounts as both of these essential components use a common metabolic precursor. Hence, checkpoints are in place, right from the regulation of the first committed step in LPS biosynthesis mediated by LpxC through its turnover by FtsH and HslUV proteases in coordination with LPS assembly factors LapB and LapC. After the synthesis of LPS on the inner leaflet of the inner membrane (IM), LPS is flipped by the IM-located essential ATP-dependent transporter to the periplasmic face of IM, where it is picked up by the LPS transport complex spanning all three components of the cell envelope for its delivery to OM. MsbA exerts its intrinsic hydrocarbon ruler function as another checkpoint to transport hexa-acylated LPS as compared to underacylated LPS. Additional checkpoints in LPS assembly are: LapB-assisted coupling of LPS synthesis and translocation; cardiolipin presence when LPS is underacylated; the recruitment of RfaH transcriptional factor ensuring the transcription of LPS core biosynthetic genes; and the regulated incorporation of non-stoichiometric modifications, controlled by the stress-responsive RpoE sigma factor, small RNAs and two-component systems.
Collapse
|
5
|
Ghannoum MA, Ford M, Bonomo RA, Gamal A, McCormick TS. A Microbiome-Driven Approach to Combating Depression During the COVID-19 Pandemic. Front Nutr 2021; 8:672390. [PMID: 34504858 PMCID: PMC8421528 DOI: 10.3389/fnut.2021.672390] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
The significant stressors brought about and exacerbated by COVID-19 are associated with startling surges in mental health illnesses, specifically those related to depressive disorders. Given the huge impact of depression on society, and an incomplete understanding of impactful therapeutics, we have examined the current literature surrounding the microbiome and gut-brain axis to advance a potential complementary approach to address depression and depressive disorders that have increased during the COVID-19 pandemic. While we understand that the impact of the human gut microbiome on emotional health is a newly emerging field and more research needs to be conducted, the current evidence is extremely promising and suggests at least part of the answer to understanding depression in more depth may lie within the microbiome. As a result of these findings, we propose that a microbiome-based holistic approach, which involves carefully annotating the microbiome and potential modification through diet, probiotics, and lifestyle changes, may address depression. This paper's primary purpose is to shed light on the link between the gut microbiome and depression, including the gut-brain axis and propose a holistic approach to microbiome modification, with the ultimate goal of assisting individuals to manage their battle with depression through diet, probiotics, and lifestyle changes, in addition to offering a semblance of hope during these challenging times.
Collapse
Affiliation(s)
- Mahmoud A. Ghannoum
- Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- BIOHM Health LLC, Cleveland, OH, United States
| | | | - Robert A. Bonomo
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Ahmed Gamal
- Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Thomas S. McCormick
- Integrated Microbiome Core, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
6
|
Viennois E, Bretin A, Dubé PE, Maue AC, Dauriat CJG, Barnich N, Gewirtz AT, Chassaing B. Dietary Emulsifiers Directly Impact Adherent-Invasive E. coli Gene Expression to Drive Chronic Intestinal Inflammation. Cell Rep 2020; 33:108229. [PMID: 33027647 PMCID: PMC7539532 DOI: 10.1016/j.celrep.2020.108229] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/30/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Dietary emulsifiers carboxymethylcellulose (CMC) and polysorbate-80 (P80) disturb gut microbiota, promoting chronic inflammation. Mice with minimal microbiota are protected against emulsifiers’ effects, leading us to hypothesize that these compounds might provoke select pathobionts to promote inflammation. Gnotobiotic wild-type (WT) and interleukin-10 (IL-10)−/− mice were colonized with Crohn’s-disease-associated adherent-invasive E. coli (AIEC) and subsequently administered CMC or P80. AIEC colonization of GF and altered Schaedler flora (ASF) mice results in chronic intestinal inflammation and metabolism dysregulations when consuming the emulsifier. In IL-10−/− mice, AIEC mono-colonization results in severe intestinal inflammation in response to emulsifiers. Exposure of AIEC to emulsifiers in vitro increases its motility and ability to adhere to intestinal epithelial cells. Transcriptomic analysis reveals that emulsifiers directly induce expression of clusters of genes that mediate AIEC virulence and promotion of inflammation. To conclude, emulsifiers promote virulence and encroachment of pathobionts, providing a means by which these compounds may drive inflammation in hosts carrying such bacteria. Dietary emulsifiers alter the intestinal microbiota, promoting chronic inflammation Select pathobionts are required to mediate the detrimental effects of emulsifiers Emulsifiers directly induce the expression of bacterial virulence genes Microbiota-based dietary intervention appears warranted
Collapse
Affiliation(s)
- Emilie Viennois
- INSERM, U1149, Center of Research on Inflammation, Université de Paris, Paris, France; Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Alexis Bretin
- Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | | | | | - Charlène J G Dauriat
- INSERM U1016, team "Mucosal microbiota in chronic inflammatory diseases", CNRS UMR 8104, Université de Paris, Paris, France
| | - Nicolas Barnich
- Université Clermont Auvergne/Inserm U1071 USC-INRA 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), Clermont-Ferrand, France
| | - Andrew T Gewirtz
- Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Benoit Chassaing
- Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA; INSERM U1016, team "Mucosal microbiota in chronic inflammatory diseases", CNRS UMR 8104, Université de Paris, Paris, France; Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
7
|
Liang H, Zhang Y, Wang S, Gao H. Mutual interplay between ArcA and σ E orchestrates envelope stress response in Shewanella oneidensis. Environ Microbiol 2020; 23:652-668. [PMID: 32372525 DOI: 10.1111/1462-2920.15060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/03/2020] [Indexed: 01/15/2023]
Abstract
To survive and thrive in harsh and ever-changing environments, intricate mechanisms have evolved for bacterial cells to monitor perturbations impacting the integrity of their envelope and to mount an appropriate response to contain or repair the damage. In this study, we report in Shewanella oneidensis a previously undescribed mechanism for the envelope defect resulting from the loss of Arc, a two-component transcriptional regulatory system crucial for respiration. We uncovered σE , a master regulator establishing and maintaining the integrity of the cell envelope in γ-proteobacteria, as the determining factor for the cell envelope defect of the arcA mutant. When ArcA is depleted, σE activity is compromised by enhanced production of anti-σE protein RseA. Surprisingly, S. oneidensis σE is not essential for viability, but becomes so in the absence of ArcA. Furthermore, we demonstrated that there is an interplay between these two regulators as arcA expression is affected by availability of σE . Overall, our results underscore functional interplay of regulatory systems for envelope stress response: although each of the systems may respond to perturbation of particular components of the envelope, they are functionally intertwined, working together to form an interconnected safety net.
Collapse
Affiliation(s)
- Huihui Liang
- Institute of Microbiology and College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| | - Yongting Zhang
- Institute of Microbiology and College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| | - Sijing Wang
- Institute of Microbiology and College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| | - Haichun Gao
- Institute of Microbiology and College of Life Sciences, Zhejiang University, Zhejiang, Hangzhou, 310058, China
| |
Collapse
|
8
|
Abstract
The effects of diversity of the gut microbiome on inflammation have centered mainly on bacterial flora. Recent research has implicated fungal species and their interactions with other organisms in the inflammatory process. New ways to restore microbial balance in the gut are being explored. Our goal was to identify beneficial probiotic strains that would antagonize these fungal and bacterial pathogens that are elevated in the inflamed gut, and which also have antibiofilm activity. Fungus-bacterium correlation analysis allowed us to identify candidate probiotic species that can antagonize microbial pathogens, which we subsequently incorporated into a novel probiotic formulation. Amylase, which is known to have some antibiofilm activity, was also added to the probiotic mixture. This novel probiotic may have utility for the management of inflammatory bowel diseases by disrupting polymicrobial biofilm formation. Dysbiosis of the gut microbiome has been implicated in inflammatory bowel diseases. We have shown that levels of Candida tropicalis, along with those of Escherichia coli and Serratia marcescens, are significantly elevated in Crohn’s disease (CD) patients. Here, we evaluated the ability of a novel probiotic to prevent and treat polymicrobial biofilms (PMB) formed by C. tropicalis with E. coli and S. marcescens. Since Candida albicans has been reported to be elevated in CD patients, we investigated the interactions of C. albicans with these bacterial species in biofilm formation. We determined whether the interaction between Candida spp. and bacteria is specific by using Trichosporon inkin and Saccharomyces fibuligera as comparators. Additionally, the effects of probiotics on C. albicans germination and biofilm formation were determined. To determine the ability of the probiotic to prevent or treat mature biofilms, probiotic filtrate was added to the PMB at early (prevention) and mature (treatment) phases. Biofilm thickness and architecture were assessed by confocal scanning laser microscopy. The effects of the probiotic on germination were evaluated in the presence of serum. Exposure of C. tropicalis PMB to probiotic filtrate reduced biofilm matrix, decreased thickness, and inhibited hyphal formation. We showed that C. albicans or C. tropicalis formed significantly thicker PMB than control biofilms, indicating that this interaction is Candida specific. Treatment with probiotic filtrate inhibited C. albicans germination and prevented/treated C. albicans PMB. The designed probiotic may have utility in the management of biofilm-associated gastrointestinal diseases such as Crohn’s and colorectal cancer.
Collapse
|
9
|
Chen CC, Chen YN, Liou JM, Wu MS. From germ theory to germ therapy. Kaohsiung J Med Sci 2019; 35:73-82. [DOI: 10.1002/kjm2.12011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/22/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Chieh-Chang Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine; National Taiwan University Hospital; Taipei Taiwan
- Department of Internal Medicine, College of Medicine; National Taiwan University; Taipei Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine; National Taiwan University; Taipei Taiwan
| | - Yen-Nien Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine; National Taiwan University Hospital; Taipei Taiwan
- Department of Internal Medicine, College of Medicine; National Taiwan University; Taipei Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine; National Taiwan University; Taipei Taiwan
| | - Jyh-Ming Liou
- Division of Gastroenterology and Hepatology, Department of Internal Medicine; National Taiwan University Hospital; Taipei Taiwan
- Department of Internal Medicine, College of Medicine; National Taiwan University; Taipei Taiwan
| | - Ming-Shiang Wu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine; National Taiwan University Hospital; Taipei Taiwan
- Department of Internal Medicine, College of Medicine; National Taiwan University; Taipei Taiwan
| | | |
Collapse
|
10
|
Renouf MJ, Cho YH, McPhee JB. Emergent Behavior of IBD-Associated Escherichia coli During Disease. Inflamm Bowel Dis 2019; 25:33-44. [PMID: 30321333 DOI: 10.1093/ibd/izy312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel diseases are becoming increasingly common throughout the world, both in developed countries and increasingly in rapidly developing countries. Multiple lines of evidence point to a role for the microbial composition of the gastrointestinal tract in the etiology of IBD, but to date, attempts to define a specific microbial cause for IBD have proved unsuccessful. Microbial 16S rRNA profiling shows that IBD patients have elevated levels of Enterobacteriaceae, in particular Escherichia coli, and reduced levels of Faecalibacterium prausnitzii. The observed E. coli have been assigned to a specific pathovar, adherent-invasive E. coli (AIEC). Adherent-invasive E. coli are a genomically heterogenous group, and whereas many groups have attempted to identify specific genetic markers that differentiate AIEC from non-AIEC strains, very few concrete genetic associations have been uncovered. Here, we highlight the advantages of applying a phenotyping approach to the study of these organisms, rather than solely depending on a sequencing or genomic-based screening strategy because virulence-associated phenotypes exhibit behaviors of emergent systems. In this respect, attempts at genetic reductionism are prone to failure because there are numerous metabolic, regulatory or genetic paths that can underlie these virulence-associated behaviors. Here, we review these IBD-associated phenotypes in E. coli and make recommendations for experimental approaches to advance our understanding of IBD-associated bacteria more generally. With advances in high-throughput screening and nongenetically based metabolomic characterization of IBD-associated bacteria, we anticipate a fuller understanding of how altered microbial communities contribute to the development of IBD.
Collapse
Affiliation(s)
| | - Youn Hee Cho
- Department of Chemistry and Biology, Ryerson University, Toronto ON, Canada
| | - Joseph B McPhee
- Department of Chemistry and Biology, Ryerson University, Toronto ON, Canada
| |
Collapse
|
11
|
Palmela C, Chevarin C, Xu Z, Torres J, Sevrin G, Hirten R, Barnich N, Ng SC, Colombel JF. Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut 2018; 67:574-587. [PMID: 29141957 DOI: 10.1136/gutjnl-2017-314903] [Citation(s) in RCA: 329] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/20/2017] [Accepted: 10/28/2017] [Indexed: 02/06/2023]
Abstract
Intestinal microbiome dysbiosis has been consistently described in patients with IBD. In the last decades, Escherichia coli, and the adherent-invasive E coli (AIEC) pathotype in particular, has been implicated in the pathogenesis of IBD. Since the discovery of AIEC, two decades ago, progress has been made in unravelling these bacteria characteristics and its interaction with the gut immune system. The mechanisms of adhesion of AIEC to intestinal epithelial cells (via FimH and cell adhesion molecule 6) and its ability to escape autophagy when inside macrophages are reviewed here. We also explore the existing data on the prevalence of AIEC in patients with Crohn's disease and UC, and the association between the presence of AIEC and disease location, activity and postoperative recurrence. Finally, we highlight potential therapeutic strategies targeting AIEC colonisation of gut mucosa, including the use of phage therapy, bacteriocins and antiadhesive molecules. These strategies may open new avenues for the prevention and treatment of IBD in the future.
Collapse
Affiliation(s)
- Carolina Palmela
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Caroline Chevarin
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Zhilu Xu
- Department of Medicine and Therapeutics, Institute of Digestive Diseases, LKS Institute of Health Science, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Joana Torres
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Gwladys Sevrin
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Robert Hirten
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Nicolas Barnich
- Université Clermont Auvergne, Inserm U1071, USC-INRA 2018, M2iSH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - Siew C Ng
- Department of Medicine and Therapeutics, Institute of Digestive Diseases, LKS Institute of Health Science, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
12
|
Zhang L, Carmody RN, Kalariya HM, Duran RM, Moskal K, Poulev A, Kuhn P, Tveter KM, Turnbaugh PJ, Raskin I, Roopchand DE. Grape proanthocyanidin-induced intestinal bloom of Akkermansia muciniphila is dependent on its baseline abundance and precedes activation of host genes related to metabolic health. J Nutr Biochem 2018; 56:142-151. [PMID: 29571008 DOI: 10.1016/j.jnutbio.2018.02.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/24/2018] [Accepted: 02/08/2018] [Indexed: 12/12/2022]
Abstract
We previously showed that C57BL/6J mice fed high-fat diet (HFD) supplemented with 1% grape polyphenols (GP) for 12 weeks developed a bloom of Akkermansia muciniphila with attenuated metabolic syndrome symptoms. Here we investigated early timing of GP-induced effects and the responsible class of grape polyphenols. Mice were fed HFD, low-fat diet (LFD) or formulations supplemented with GP (HFD-GP, LFD-GP) for 14 days. Mice fed HFD-GP, but not LFD-GP, showed improved oral glucose tolerance compared to controls. A. muciniphila bloom occurred earlier in mice fed LFD-GP than HFD-GP; however, timing was dependent on baseline A. muciniphila levels rather than dietary fat. Mice gavaged for 10 days with GP extract (GPE) or grape proanthocyanidins (PACs), each delivering 360 mg PACs/kg body weight, induced a bloom of fecal and cecal A. muciniphila, the rate of which depended on initial A. muciniphila abundance. Grape PACs were sufficient to induce a bloom of A. muciniphila independent of specific intestinal gene expression changes. Gut microbial community analysis and in vitro inhibition of A. muciniphila by GPE or PACs suggest that the A. muciniphila bloom in vivo occurs via indirect mechanisms.
Collapse
Affiliation(s)
- Li Zhang
- Rutgers, The State University of New Jersey, Department of Plant Biology, Foran Hall, 59 Dudley Road, New Brunswick, NJ 08901, USA
| | - Rachel N Carmody
- Harvard University, Department of Human Evolutionary Biology, 11 Divinity Avenue, Cambridge, MA 02138
| | - Hetal M Kalariya
- Rutgers, The State University of New Jersey, Department of Plant Biology, Foran Hall, 59 Dudley Road, New Brunswick, NJ 08901, USA
| | - Rocio M Duran
- Rutgers, The State University of New Jersey, Department of Food Science, Institute for Food Nutrition and Health, Center for Digestive Health, 61 Dudley Road, New Brunswick, NJ 08901, USA
| | - Kristin Moskal
- Rutgers, The State University of New Jersey, Department of Plant Biology, Foran Hall, 59 Dudley Road, New Brunswick, NJ 08901, USA
| | - Alexander Poulev
- Rutgers, The State University of New Jersey, Department of Plant Biology, Foran Hall, 59 Dudley Road, New Brunswick, NJ 08901, USA
| | - Peter Kuhn
- Rutgers, The State University of New Jersey, Department of Plant Biology, Foran Hall, 59 Dudley Road, New Brunswick, NJ 08901, USA
| | - Kevin M Tveter
- Rutgers, The State University of New Jersey, Department of Food Science, Institute for Food Nutrition and Health, Center for Digestive Health, 61 Dudley Road, New Brunswick, NJ 08901, USA
| | - Peter J Turnbaugh
- University of California San Francisco, Department of Microbiology & Immunology, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Ilya Raskin
- Rutgers, The State University of New Jersey, Department of Plant Biology, Foran Hall, 59 Dudley Road, New Brunswick, NJ 08901, USA
| | - Diana E Roopchand
- Rutgers, The State University of New Jersey, Department of Food Science, Institute for Food Nutrition and Health, Center for Digestive Health, 61 Dudley Road, New Brunswick, NJ 08901, USA.
| |
Collapse
|
13
|
O'Brien CL, Bringer MA, Holt KE, Gordon DM, Dubois AL, Barnich N, Darfeuille-Michaud A, Pavli P. Comparative genomics of Crohn's disease-associated adherent-invasive Escherichia coli. Gut 2017; 66:1382-1389. [PMID: 27196580 DOI: 10.1136/gutjnl-2015-311059] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 03/22/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Adherent-invasive Escherichia coli (AIEC) are a leading candidate bacterial trigger for Crohn's disease (CD). The AIEC pathovar is defined by in vitro cell-line assays examining specific bacteria/cell interactions. No molecular marker exists for their identification. Our aim was to identify a molecular property common to the AIEC phenotype. DESIGN 41 B2 phylogroup E. coli strains were isolated from 36 Australian subjects: 19 patients with IBD and 17 without. Adherence/invasion assays were conducted using the I-407 epithelial cell line and survival/replication assays using the THP-1 macrophage cell line. Cytokine secretion tumour necrosis factor ((TNF)-α, interleukin (IL) 6, IL-8 and IL-10) was measured using ELISA. The genomes were assembled and annotated, and cluster analysis performed using CD-HIT. The resulting matrices were analysed to identify genes unique/more frequent in AIEC strains compared with non-AIEC strains. Base composition differences and clustered regularly interspaced palindromic repeat (CRISPR) analyses were conducted. RESULTS Of all B2 phylogroup strains assessed, 79% could survive and replicate in macrophages. Among them, 11/41 strains (5 CD, 2 UCs, 5 non-IBD) also adhere to and invade epithelial cells, a phenotype assigning them to the AIEC pathovar. The AIEC strains were phylogenetically heterogeneous. We did not identify a gene (or nucleic acid base composition differences) common to all, or the majority of, AIEC. Cytokine secretion and CRISPRs were not associated with the AIEC phenotype. CONCLUSIONS Comparative genomic analysis of AIEC and non-AIEC strains did not identify a molecular property exclusive to the AIEC phenotype. We recommend a broader approach to the identification of the bacteria-host interactions that are important in the pathogenesis of Crohn's disease.
Collapse
Affiliation(s)
- Claire L O'Brien
- Medical School, Australian National University, Canberra, Australian Capital Territory, Australia.,Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Marie-Agnès Bringer
- INRA UMR1324, CNRS UMR6265, Université Bourgogne-Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, Dijon, France.,UMR1071 Inserm/University of Auvergne, INRA USC2018, M2iSH, Clermont-Ferrand, France
| | - Kathryn E Holt
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - David M Gordon
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Anaëlle L Dubois
- UMR1071 Inserm/University of Auvergne, INRA USC2018, M2iSH, Clermont-Ferrand, France
| | - Nicolas Barnich
- UMR1071 Inserm/University of Auvergne, INRA USC2018, M2iSH, Clermont-Ferrand, France
| | | | - Paul Pavli
- Medical School, Australian National University, Canberra, Australian Capital Territory, Australia.,Gastroenterology and Hepatology Unit, Canberra Hospital, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
14
|
Sartor RB, Wu GD. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology 2017; 152:327-339.e4. [PMID: 27769810 PMCID: PMC5511756 DOI: 10.1053/j.gastro.2016.10.012] [Citation(s) in RCA: 556] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 02/08/2023]
Abstract
Intestinal microbiota are involved in the pathogenesis of Crohn's disease, ulcerative colitis, and pouchitis. We review the mechanisms by which these gut bacteria, fungi, and viruses mediate mucosal homeostasis via their composite genes (metagenome) and metabolic products (metabolome). We explain how alterations to their profiles and functions under conditions of dysbiosis contribute to inflammation and effector immune responses that mediate inflammatory bowel diseases (IBD) in humans and enterocolitis in mice. It could be possible to engineer the intestinal environment by modifying the microbiota community structure or function to treat patients with IBD-either with individual agents, via dietary management, or as adjuncts to immunosuppressive drugs. We summarize the latest information on therapeutic use of fecal microbial transplantation and propose improved strategies to selectively normalize the dysbiotic microbiome in personalized approaches to treatment.
Collapse
Affiliation(s)
- R Balfour Sartor
- Departments of Medicine, Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - Gary D Wu
- Division of Gastroenterology, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
15
|
The Adherent/Invasive Escherichia coli Strain LF82 Invades and Persists in Human Prostate Cell Line RWPE-1, Activating a Strong Inflammatory Response. Infect Immun 2016; 84:3105-3113. [PMID: 27600504 PMCID: PMC5067744 DOI: 10.1128/iai.00438-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/30/2016] [Indexed: 01/26/2023] Open
Abstract
Adherent/invasive Escherichia coli (AIEC) strains have recently been receiving increased attention because they are more prevalent and persistent in the intestine of Crohn's disease (CD) patients than in healthy subjects. Since AIEC strains show a high percentage of similarity to extraintestinal pathogenic E. coli (ExPEC), neonatal meningitis-associated E. coli (NMEC), and uropathogenic E. coli (UPEC) strains, here we compared AIEC strain LF82 with a UPEC isolate (strain EC73) to assess whether LF82 would be able to infect prostate cells as an extraintestinal target. The virulence phenotypes of both strains were determined by using the RWPE-1 prostate cell line. The results obtained indicated that LF82 and EC73 are able to adhere to, invade, and survive within prostate epithelial cells. Invasion was confirmed by immunofluorescence and electron microscopy. Moreover, cytochalasin D and colchicine strongly inhibited bacterial uptake of both strains, indicating the involvement of actin microfilaments and microtubules in host cell invasion. Moreover, both strains belong to phylogenetic group B2 and are strong biofilm producers. In silico analysis reveals that LF82 shares with UPEC strains several virulence factors: namely, type 1 pili, the group II capsule, the vacuolating autotransporter toxin, four iron uptake systems, and the pathogenic island (PAI). Furthermore, compared to EC73, LF82 induces in RWPE-1 cells a marked increase of phosphorylation of mitogen-activated protein kinases (MAPKs) and of NF-κB already by 5 min postinfection, thus inducing a strong inflammatory response. Our in vitro data support the hypothesis that AIEC strains might play a role in prostatitis, and, by exploiting host-cell signaling pathways controlling the innate immune response, likely facilitate bacterial multiplication and dissemination within the male genitourinary tract.
Collapse
|
16
|
Dysbiosis in intestinal inflammation: Cause or consequence. Int J Med Microbiol 2016; 306:302-309. [PMID: 27012594 DOI: 10.1016/j.ijmm.2016.02.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 02/26/2016] [Accepted: 02/29/2016] [Indexed: 02/07/2023] Open
Abstract
The intestinal microbiota encompasses hundreds of bacterial species that constitute a relatively stable ecosystem. Alteration in the microbiota composition may arise from infections, immune defects, metabolic alterations, diet or antibiotic treatment. Dysbiosis is considered as an alteration in microbiota community structure and/or function, capable of causing/driving a detrimental distortion of microbe-host homeostasis. A variety of pathologies are associated with changes in the community structure and function of the gut microbiota, suggesting a link between dysbiosis and disease etiology. With an emphasis in this review on inflammatory bowel diseases (IBD), the non-trivial question is whether dysbiosis is the cause or consequence of inflammation. It is important to understand whether changes in microbial ecosystems are causally linked to the pathology and to what extend disease risk is predicable based on characteristic changes in community structure and/or function. Local changes in tissue integrity associated with focal areas of inflammation may result in the selection of a dysbiotic bacterial community associated with the propagation of a disease phenotype. This review outlines the role of dysbiosis in intestinal inflammation with particular focus on IBD-relevant gnotobiotic mouse models, the factors implicated in the development of dysbiosis and the means available to investigate dysbiosis in the context of human diseases.
Collapse
|
17
|
Dahdouh E, Orgaz B, Gómez-Gil R, Mingorance J, Daoud Z, Suarez M, San Jose C. Patterns of biofilm structure and formation kinetics among Acinetobacter baumannii clinical isolates with different antibiotic resistance profiles. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00377f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study evaluates the rates of biofilm formation in light of the different characteristics of twelve A. baumannii clinical isolates.
Collapse
Affiliation(s)
- E. Dahdouh
- Faculty of Veterinary
- Department of Animal Health
- University Complutense of Madrid
- Madrid
- Spain
| | - B. Orgaz
- Faculty of Veterinary
- Department of Food Science and Technology
- University Complutense of Madrid
- Madrid
- Spain
| | - R. Gómez-Gil
- Servicio de Microbiología
- Hospital Universitario La Paz
- IdiPAZ
- Madrid
- Spain
| | - J. Mingorance
- Servicio de Microbiología
- Hospital Universitario La Paz
- IdiPAZ
- Madrid
- Spain
| | - Z. Daoud
- Faculty of Medicine and Medical Sciences
- Department of Clinical Microbiology
- University of Balamand
- Amioun
- Lebanon
| | - M. Suarez
- Faculty of Veterinary
- Department of Animal Health
- University Complutense of Madrid
- Madrid
- Spain
| | - C. San Jose
- Faculty of Veterinary
- Department of Food Science and Technology
- University Complutense of Madrid
- Madrid
- Spain
| |
Collapse
|
18
|
Surface Polysaccharide Mutants Reveal that Absence of O Antigen Reduces Biofilm Formation of Actinobacillus pleuropneumoniae. Infect Immun 2015; 84:127-37. [PMID: 26483403 DOI: 10.1128/iai.00912-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/12/2015] [Indexed: 12/24/2022] Open
Abstract
Actinobacillus pleuropneumoniae is a Gram-negative bacterium belonging to the Pasteurellaceae family and the causative agent of porcine pleuropneumonia, a highly contagious lung disease causing important economic losses. Surface polysaccharides, including lipopolysaccharides (LPS) and capsular polysaccharides (CPS), are implicated in the adhesion and virulence of A. pleuropneumoniae, but their role in biofilm formation is still unclear. In this study, we investigated the requirement for these surface polysaccharides in biofilm formation by A. pleuropneumoniae serotype 1. Well-characterized mutants were used: an O-antigen LPS mutant, a truncated core LPS mutant with an intact O antigen, a capsule mutant, and a poly-N-acetylglucosamine (PGA) mutant. We compared the amount of biofilm produced by the parental strain and the isogenic mutants using static and dynamic systems. Compared to the findings for the biofilm of the parental or other strains, the biofilm of the O antigen and the PGA mutants was dramatically reduced, and it had less cell-associated PGA. Real-time PCR analyses revealed a significant reduction in the level of pgaA, cpxR, and cpxA mRNA in the biofilm cells of the O-antigen mutant compared to that in the biofilm cells of the parental strain. Specific binding between PGA and LPS was consistently detected by surface plasmon resonance, but the lack of O antigen did not abolish these interactions. In conclusion, the absence of the O antigen reduces the ability of A. pleuropneumoniae to form a biofilm, and this is associated with the reduced expression and production of PGA.
Collapse
|