1
|
Panpetch W, Tumwasorn S, Leelahavanichkul A. Presence of Pseudomonas aeruginosa in feces exacerbate leaky gut in mice with low dose dextran sulfate solution, impacts of specific bacteria. PLoS One 2024; 19:e0309106. [PMID: 39546435 PMCID: PMC11567622 DOI: 10.1371/journal.pone.0309106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 08/05/2024] [Indexed: 11/17/2024] Open
Abstract
The impact of Pseudomonas aeruginosa (PA) was explored in a mouse model with non-diarrheal gut permeability defect using 1.5% dextran sulfate solution (DSS) plus antibiotics (ATB) with or without orally administered PA. As such, ATB+DSS+PA mice induced more severe intestinal injury as indicated by stool consistency and leaky gut (FITC-dextran assay, bacteremia, and endotoxemia) with an increase in serum cytokines, liver enzyme, and hepatocyte apoptosis when compared with ATB+DSS mice. There was no abnormality by these parameters in the non-DSS group, including water alone (control), antibiotics alone (ATB+water), and antibiotics with PA (ATB+water+PA). Despite a similarly fecal microbiome patterns between ATB+DSS and ATB+DSS+PA groups, a higher abundance of Pseudomonas, Enterococci, and Escherichia-Shigella was detected in ATB+DSS+PA mice. Additionally, the additive pro-inflammation between pathogen molecules, using heat-killed P. aeruginosa preparations, and LPS against enterocytes (Caco2) and hepatocytes (HegG2), as indicated by supernatant IL-8 and expression of several genes (IL-8, NF-kB, and NOS2) are demonstrated. In conclusion, presence of P. aeruginosa in the gut exacerbated DSS-induced intestinal injury with spontaneous translocation of LPS and bacteria from the gut into the blood circulation (leaky gut) that induced more severe systemic inflammation. The presence of pathogenic bacteria, especially PA in stool of the healthy individuals might have some adverse effect. More studies are in needed.
Collapse
Affiliation(s)
- Wimonrat Panpetch
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Science, Burapha University, Chonburi, Thailand
| | - Somying Tumwasorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Translational Research on Immunology and Immune-Mediated Diseases (CETRII), Department of Microbiology, Faculty of Medicine, Bangkok, Thailand
| |
Collapse
|
2
|
Bravo Iniguez A, Sun Q, Cui Q, Du M, Zhu MJ. Cannabidiol Enhances Mitochondrial Metabolism and Antioxidant Defenses in Human Intestinal Epithelial Caco-2 Cells. Nutrients 2024; 16:3843. [PMID: 39599629 PMCID: PMC11597683 DOI: 10.3390/nu16223843] [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: 10/23/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The reintroduction of hemp production has resulted in increased consumption of cannabidiol (CBD) products, particularly CBD oil, yet their effects on intestinal health are not fully understood. Proper mitochondrial function and antioxidant defenses are vital for maintaining the intestinal epithelial barrier. AMP-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator (PGC)1α are key mediators of mitochondrial metabolism. METHODS & RESULTS Using Caco-2 cells, we found that CBD oil promoted AMPK phosphorylation, upregulated differentiation markers, and enhanced PGC1α/SIRT3 mitochondrial signaling. CBD oil reduced reactive oxygen species production and increased antioxidant enzymes. Moreover, CBD oil also increased levels of citrate, malate, and succinate-key metabolites of the tricarboxylic acid cycle-alongside upregulation of pyruvate dehydrogenase and isocitrate dehydrogenase 1. Similarly, pure CBD induced metabolic and antioxidant signaling. CONCLUSIONS CBD enhances mitochondrial metabolic activity and antioxidant defense in Caco-2 cells, making it a promising candidate for treating intestinal dysfunction.
Collapse
Affiliation(s)
- Alejandro Bravo Iniguez
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| | - Qi Sun
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| | - Qiaorong Cui
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA;
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA; (A.B.I.); (Q.S.); (Q.C.)
| |
Collapse
|
3
|
Kawasaki M, Ambrosini YM. Differential Colonization and Mucus Ultrastructure Visualization in Bovine Ileal and Rectal Organoid-Derived Monolayers Exposed to Enterohemorrhagic Escherichia coli. Int J Mol Sci 2024; 25:4914. [PMID: 38732126 PMCID: PMC11084217 DOI: 10.3390/ijms25094914] [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: 04/02/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) is a critical public health concern due to its role in severe gastrointestinal illnesses in humans, including hemorrhagic colitis and the life-threatening hemolytic uremic syndrome. While highly pathogenic to humans, cattle, the main reservoir for EHEC, often remain asymptomatic carriers, complicating efforts to control its spread. Our study introduces a novel method to investigate EHEC using organoid-derived monolayers from adult bovine ileum and rectum. These polarized epithelial monolayers were exposed to EHEC for four hours, allowing us to perform comparative analyses between the ileal and rectal tissues. Our findings mirrored in vivo observations, showing a higher colonization rate in the rectum compared with the ileum (44.0% vs. 16.5%, p < 0.05). Both tissues exhibited an inflammatory response with increased expression levels of TNF-a (p < 0.05) and a more pronounced increase of IL-8 in the rectum (p < 0.01). Additionally, the impact of EHEC on the mucus barrier varied across these gastrointestinal regions. Innovative visualization techniques helped us study the ultrastructure of mucus, revealing a net-like mucin glycoprotein organization. While further cellular differentiation could enhance model accuracy, our research significantly deepens understanding of EHEC pathogenesis in cattle and informs strategies for the preventative measures and therapeutic interventions.
Collapse
Affiliation(s)
| | - Yoko M. Ambrosini
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA;
| |
Collapse
|
4
|
Lange ME, Clarke ST, Boras VF, Brown CLJ, Zhang G, Laing CR, Uwiera RRE, Montina T, Kalmokoff ML, Taboada EN, Gannon VPJ, Metz GAS, Church JS, Inglis GD. Commensal Escherichia coli Strains of Bovine Origin Competitively Mitigated Escherichia coli O157:H7 in a Gnotobiotic Murine Intestinal Colonization Model with or without Physiological Stress. Animals (Basel) 2023; 13:2577. [PMID: 37627368 PMCID: PMC10451813 DOI: 10.3390/ani13162577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Cattle are a primary reservoir of enterohemorrhagic Escherichia coli (EHEC) O157:H7. Currently, there are no effective methods of eliminating this important zoonotic pathogen from cattle, and colonization resistance in relation to EHEC O157:H7 in cattle is poorly understood. We developed a gnotobiotic EHEC O157:H7 murine model to examine aspects of the cattle pathogen-microbiota interaction, and to investigate competitive suppression of EHEC O157:H7 by 18 phylogenetically distinct commensal E. coli strains of bovine origin. As stress has been suggested to influence enteric colonization by EHEC O157:H7 in cattle, corticosterone administration (±) to incite a physiological stress response was included as an experimental variable. Colonization of the intestinal tract (IT) of mice by the bovine EHEC O157:H7 strain, FRIK-2001, mimicked characteristics of bovine IT colonization. In this regard, FRIK-2001 successfully colonized the IT and temporally incited minimal impacts on the host relative to other EHEC O157:H7 strains, including on the renal metabolome. The presence of the commensal E. coli strains decreased EHEC O157:H7 densities in the cecum, proximal colon, and distal colon. Moreover, histopathologic changes and inflammation markers were reduced in the distal colon of mice inoculated with commensal E. coli strains (both propagated separately and communally). Although stress induction affected the behavior of mice, it did not influence EHEC O157:H7 densities or disease. These findings support the use of a gnotobiotic murine model of enteric bovine EHEC O157:H7 colonization to better understand pathogen-host-microbiota interactions toward the development of effective on-farm mitigations for EHEC O157:H7 in cattle, including the identification of bacteria capable of competitively colonizing the IT.
Collapse
Affiliation(s)
- Maximo E. Lange
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada; (M.E.L.); (S.T.C.); (C.L.J.B.)
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Sandra T. Clarke
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada; (M.E.L.); (S.T.C.); (C.L.J.B.)
| | - Valerie F. Boras
- Chinook Regional Hospital, Alberta Health Services, Lethbridge, AB T1J 1W5, Canada;
| | - Catherine L. J. Brown
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada; (M.E.L.); (S.T.C.); (C.L.J.B.)
| | - Guangzhi Zhang
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (G.Z.); (E.N.T.)
| | - Chad R. Laing
- National Centre for Animal Diseases, Canadian Food Inspection Agency, Lethbridge, AB T1J 3Z4, Canada;
| | - Richard R. E. Uwiera
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Tony Montina
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada;
| | - Martin L. Kalmokoff
- Kentville Research and Development Centre, Agriculture and Agri-Food Canada, Kentville, NS B4N 1J5, Canada;
| | - Eduardo N. Taboada
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (G.Z.); (E.N.T.)
| | - Victor P. J. Gannon
- National Microbiology Laboratory, Public Health Agency of Canada, Lethbridge, AB T1J 3Z4, Canada;
| | - Gerlinde A. S. Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada;
| | - John S. Church
- Natural Resource Science, Thompson Rivers University, Kamloops, BC V2C 0C8, Canada;
| | - G. Douglas Inglis
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada; (M.E.L.); (S.T.C.); (C.L.J.B.)
| |
Collapse
|
5
|
Monteiro R, Chafsey I, Caccia N, Ageorges V, Leroy S, Viala D, Hébraud M, Livrelli V, Pizza M, Pezzicoli A, Desvaux M. Specific Proteomic Identification of Collagen-Binding Proteins in Escherichia coli O157:H7: Characterisation of OmpA as a Potent Vaccine Antigen. Cells 2023; 12:1634. [PMID: 37371104 DOI: 10.3390/cells12121634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Escherichia coli is a versatile commensal species of the animal gut that can also be a pathogen able to cause intestinal and extraintestinal infections. The plasticity of its genome has led to the evolution of pathogenic strains, which represent a threat to global health. Additionally, E. coli strains are major drivers of antibiotic resistance, highlighting the urgent need for new treatment and prevention measures. The antigenic and structural heterogeneity of enterohaemorrhagic E. coli colonisation factors has limited their use for the development of effective and cross-protective vaccines. However, the emergence of new strains that express virulence factors deriving from different E. coli diarrhoeagenic pathotypes suggests that a vaccine targeting conserved proteins could be a more effective approach. In this study, we conducted proteomics analysis and functional protein characterisation to identify a group of proteins potentially involved in the adhesion of E. coli O157:H7 to the extracellular matrix and intestinal epithelial cells. Among them, OmpA has been identified as a highly conserved and immunogenic antigen, playing a significant role in the adhesion phenotype of E. coli O157:H7 and in bacterial aggregation. Furthermore, antibodies raised against recombinant OmpA effectively reduced the adhesion of E. coli O157:H7 to intestinal epithelial cells. The present work highlights the role of OmpA as a potent antigen for the development of a vaccine against intestinal pathogenic E. coli.
Collapse
Affiliation(s)
- Ricardo Monteiro
- INRAE, UCA, UMR0454 MEDIS, 63000 Clermont-Ferrand, France
- GSK, 53100 Siena, Italy
- Instituto de Investigação e Inovação em Saúde-i3S, Universidade do Porto, 4150-564 Porto, Portugal
| | - Ingrid Chafsey
- INRAE, UCA, UMR0454 MEDIS, 63000 Clermont-Ferrand, France
| | - Nelly Caccia
- INRAE, UCA, UMR0454 MEDIS, 63000 Clermont-Ferrand, France
| | | | - Sabine Leroy
- INRAE, UCA, UMR0454 MEDIS, 63000 Clermont-Ferrand, France
| | - Didier Viala
- INRAE, Metabolism Exploration Platform, Proteomic Component (PFEMcp), 63122 Saint-Genès Champanelle, France
| | - Michel Hébraud
- INRAE, UCA, UMR0454 MEDIS, 63000 Clermont-Ferrand, France
- INRAE, Metabolism Exploration Platform, Proteomic Component (PFEMcp), 63122 Saint-Genès Champanelle, France
| | | | - Mariagrazia Pizza
- GSK, 53100 Siena, Italy
- Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | | | | |
Collapse
|
6
|
Gu M, Yin W, Zhang J, Yin J, Tang X, Ling J, Tang Z, Yin W, Wang X, Ni Q, Zhu Y, Chen T. Role of gut microbiota and bacterial metabolites in mucins of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1119992. [PMID: 37265504 PMCID: PMC10229905 DOI: 10.3389/fcimb.2023.1119992] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
Colorectal cancer (CRC) is a major health burden, accounting for approximately 10% of all new cancer cases worldwide. Accumulating evidence suggests that the crosstalk between the host mucins and gut microbiota is associated with the occurrence and development of CRC. Mucins secreted by goblet cells not only protect the intestinal epithelium from microorganisms and invading pathogens but also provide a habitat for commensal bacteria. Conversely, gut dysbiosis results in the dysfunction of mucins, allowing other commensals and their metabolites to pass through the intestinal epithelium, potentially triggering host responses and the subsequent progression of CRC. In this review, we summarize how gut microbiota and bacterial metabolites regulate the function and expression of mucin in CRC and novel treatment strategies for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiangjun Wang
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Qing Ni
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Yunxiang Zhu
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Tuo Chen
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| |
Collapse
|
7
|
Kim K, Song M, Liu Y, Ji P. Enterotoxigenic Escherichia coli infection of weaned pigs: Intestinal challenges and nutritional intervention to enhance disease resistance. Front Immunol 2022; 13:885253. [PMID: 35990617 PMCID: PMC9389069 DOI: 10.3389/fimmu.2022.885253] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) infection induced post-weaning diarrhea is one of the leading causes of morbidity and mortality in newly weaned pigs and one of the significant drivers for antimicrobial use in swine production. ETEC attachment to the small intestine initiates ETEC colonization and infection. The secretion of enterotoxins further disrupts intestinal barrier function and induces intestinal inflammation in weaned pigs. ETEC infection can also aggravate the intestinal microbiota dysbiosis due to weaning stress and increase the susceptibility of weaned pigs to other enteric infectious diseases, which may result in diarrhea or sudden death. Therefore, the amount of antimicrobial drugs for medical treatment purposes in major food-producing animal species is still significant. The alternative practices that may help reduce the reliance on such antimicrobial drugs and address animal health requirements are needed. Nutritional intervention in order to enhance intestinal health and the overall performance of weaned pigs is one of the most powerful practices in the antibiotic-free production system. This review summarizes the utilization of several categories of feed additives or supplements, such as direct-fed microbials, prebiotics, phytochemicals, lysozyme, and micro minerals in newly weaned pigs. The current understanding of these candidates on intestinal health and disease resistance of pigs under ETEC infection are particularly discussed, which may inspire more research on the development of alternative practices to support food-producing animals.
Collapse
Affiliation(s)
- Kwangwook Kim
- Department of Animal Science, University of California, Davis, Davis, CA, United States
| | - Minho Song
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, South Korea
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, Davis, CA, United States
- *Correspondence: Yanhong Liu, ; Peng Ji,
| | - Peng Ji
- Department of Nutrition, University of California, Davis, Davis, CA, United States
- *Correspondence: Yanhong Liu, ; Peng Ji,
| |
Collapse
|
8
|
Zhang MM, An LY, Hu WX, Li ZY, Qiang YY, Zhao BY, Han TS, Wu CC. Mechanism of endometrial MUC2 in reproductive performance in mice through PI3K/AKT signaling pathway after lipopolysaccharide treatment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 231:113177. [PMID: 35030527 DOI: 10.1016/j.ecoenv.2022.113177] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
The objective of this study was to investigate the effects of exposure to endotoxin on the reproductive performance of humans and animals in pregnancy and delivery period. Mucin is considered to play a critical role in protecting the tissue epithelium. At pregnancy period, the MUC2 expression of uterus in the High LPS group was significantly higher than that in the Control group. The glycosaminoglycans of gland cells were secreted into the uterine cavity to protect the uterus. Then, the MUC2 layer became thinner, and LPS entered the lamina propria of the uterus. The mRNA expression of tight junction proteins showed a marked drop, and morphological damage of the uterus occurred. Subsequently, the glycosaminoglycans of gland cells in the High LPS and Low LPS groups increased with the increasing LPS dose, and the damage to the endometrial epithelium was repaired in female mice at Day 5 postdelivery. A low dose of LPS activated the PI3K/AKT signaling pathways to increase the glycosaminoglycans particles, while a high dose of LPS inhibited the PI3K/AKT signaling pathway to decrease the glycosaminoglycans particles. Taken together, our results suggest that gland cells secreted glycosaminoglycans particles into the uterine cavity by exocytosis to increase the thickness of the mucus layer to protect the uterus and that this process was regulated by PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Ming-Ming Zhang
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Li-Yan An
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Wen-Xiang Hu
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Zhong-Yang Li
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Yu-Yun Qiang
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Bao-Yu Zhao
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China
| | - Tie-Suo Han
- Lanzhou Chia Tai Food CO. LTD, Lanzhou 730000, Gansu, People's Republic of China
| | - Chen-Chen Wu
- College of Animal Veterinary Medicine, Northwest A & F University, Yangling 712100, Shaanxi, People's Republic of China.
| |
Collapse
|
9
|
Panpetch W, Phuengmaung P, Cheibchalard T, Somboonna N, Leelahavanichkul A, Tumwasorn S. Lacticaseibacillus casei Strain T21 Attenuates Clostridioides difficile Infection in a Murine Model Through Reduction of Inflammation and Gut Dysbiosis With Decreased Toxin Lethality and Enhanced Mucin Production. Front Microbiol 2021; 12:745299. [PMID: 34925261 PMCID: PMC8672038 DOI: 10.3389/fmicb.2021.745299] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Clostridioides difficile is a major cause of diarrhea in patients with antibiotic administration. Lacticaseibacillus casei T21, isolated from a human gastric biopsy, was tested in a murine C. difficile infection (CDI) model and colonic epithelial cells (Caco-2 and HT-29). Daily administration of L. casei T21 [1 × 108 colony forming units (CFU)/dose] for 4 days starting at 1 day before C. difficile challenge attenuated CDI as demonstrated by a reduction in mortality rate, weight loss, diarrhea, gut leakage, gut dysbiosis, intestinal pathology changes, and levels of pro-inflammatory cytokines [interleukin (IL)-1β, tumor necrosis factor (TNF)-α, macrophage inflammatory protein 2 (MIP-2), and keratinocyte chemoattractant (KC)] in the intestinal tissue and serum. Conditioned media from L. casei T21 exerted biological activities that fight against C. difficile as demonstrated in colonic epithelial cells by the following: (i) suppression of gene expression and production of IL-8, an important chemokine involved in C. difficile pathogenesis, (ii) reduction in the expression of SLC11A1 (solute carrier family 11 member 1) and HuR (human antigen R), important genes for the lethality of C. difficile toxin B, (iii) augmentation of intestinal integrity, and (iv) up-regulation of MUC2, a mucosal protective gene. These results supported the therapeutic potential of L. casei T21 for CDI and the need for further study on the intervention capabilities of CDI.
Collapse
Affiliation(s)
- Wimonrat Panpetch
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thanya Cheibchalard
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Naraporn Somboonna
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Microbiome Research Unit for Probiotics in Food and Cosmetics, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, Bangkok, Thailand
- *Correspondence: Asada Leelahavanichkul,
| | - Somying Tumwasorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Microbiome Research Unit for Probiotics in Food and Cosmetics, Chulalongkorn University, Bangkok, Thailand
- Somying Tumwasorn,
| |
Collapse
|
10
|
Sun Q, Du M, Navarre DA, Zhu M. Effect of Cooking Methods on Bioactivity of Polyphenols in Purple Potatoes. Antioxidants (Basel) 2021; 10:antiox10081176. [PMID: 34439424 PMCID: PMC8388894 DOI: 10.3390/antiox10081176] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/03/2023] Open
Abstract
Purple-fleshed potato (Solanum tuberosum L.) is a good dietary source of anthocyanins, flavonols, and polyphenolic acids, mostly chlorogenic acid. The objective of this study was to examine the impacts of cooking methods including boiling, steaming, and the newly developed vacuum-sealed boiling (VSBoil) on extractability and bioactivity of polyphenolic compounds in a purple potato (PP) cultivar, Purple Pelisse. Data showed that boiling and steaming reduced the total polyphenolic content in PP. High-performance liquid chromatography analysis showed that steaming slightly reduced the extractable chlorogenic acid content, while VSBoil increased it. For DPPH free radical scavenging activities, VSBoil and steaming effectively preserved the antioxidant activity of a polyphenol-rich extract of PP, while boiling resulted in a significant reduction compared to raw potato extract. All extracts effectively suppressed bursts of intracellular reactive oxygen species in human colonic epithelial cells upon hydrogen peroxide induction, of which the extract from the VSBoil group showed the highest antioxidant potential. In addition, all extracts showed anti-inflammatory effects in Caco-2 cells induced with tumor necrosis factor-α. In conclusion, the content and bioactivity of extractable polyphenols were largely retained in PP subjected to different cooking processes. VSBoil resulted in the highest content of extractable polyphenolic compounds and bioactivity among tested cooking methods.
Collapse
Affiliation(s)
- Qi Sun
- School of Food Science, Washington State University, Pullman, WA 99164, USA;
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, WA 99164, USA;
| | - Duroy A. Navarre
- Temperate Tree Fruit and Vegetable Research Unit, USDA-Agricultural Research Service, Prosser, WA 99350, USA;
| | - Meijun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA;
- Correspondence: ; Tel.: +1-(509)-335-4016
| |
Collapse
|
11
|
Xi D, Hofmann L, Alter T, Einspanier R, Bereswill S, Heimesaat MM, Gölz G, Sharbati S. The glycosyltransferase ST3GAL2 is regulated by miR-615-3p in the intestinal tract of Campylobacter jejuni infected mice. Gut Pathog 2021; 13:42. [PMID: 34183045 PMCID: PMC8240225 DOI: 10.1186/s13099-021-00437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/10/2021] [Indexed: 11/30/2022] Open
Abstract
Background Campylobacter jejuni (C. jejuni) infections are of increasing importance worldwide. As a typical mucosal pathogen, the interaction of C. jejuni with mucins is a prominent step in the colonisation of mucosal surfaces. Despite recent advances in understanding the interaction between bacterial pathogens and host mucins, the mechanisms of mucin glycosylation during intestinal C. jejuni infection remain largely unclear. This prompted us to identify relevant regulatory networks that are concerted by miRNAs and could play a role in the mucin modification and interaction. Results We firstly used a human intestinal in vitro model, in which we observed altered transcription of MUC2 and TFF3 upon C. jejuni NCTC 11168 infection. Using a combined approach consisting of in silico analysis together with in vitro expression analysis, we identified the conserved miRNAs miR-125a-5p and miR-615-3p associated with MUC2 and TFF3. Further pathway analyses showed that both miRNAs appear to regulate glycosyltransferases, which are related to the KEGG pathway ‘Mucin type O-glycan biosynthesis’. To validate the proposed interactions, we applied an in vivo approach utilising a well-established secondary abiotic IL-10−/− mouse model for infection with C. jejuni 81-176. In colonic tissue samples, we confirmed infection-dependent aberrant transcription of MUC2 and TFF3. Moreover, two predicted glycosyltransferases, the sialyltransferases ST3GAL1 and ST3GAL2, exhibited inversely correlated transcriptional levels compared to the expression of the identified miRNAs miR-125a-5p and miR-615-3p, respectively. In this study, we mainly focused on the interaction between miR-615-3p and ST3GAL2 and were able to demonstrate their molecular interaction using luciferase reporter assays and RNAi. Detection of ST3GAL2 in murine colonic tissue by immunofluorescence demonstrated reduced intensity after C. jejuni 81-176 infection and was thus consistent with the observations made above. Conclusions We report here for the first time the regulation of glycosyltransferases by miRNAs during murine infection with C. jejuni 81-176. Our data suggest that mucin type O-glycan biosynthesis is concerted by the interplay of miRNAs and glycosyltransferases, which could determine the shape of intestinal glycosylated proteins during infection. Supplementary Information The online version contains supplementary material available at 10.1186/s13099-021-00437-1.
Collapse
Affiliation(s)
- De Xi
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Lukas Hofmann
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Thomas Alter
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Stefan Bereswill
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Markus M Heimesaat
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Greta Gölz
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Stromberg ZR, Masonbrink RE, Mellata M. Transcriptomic Analysis of Shiga Toxin-Producing Escherichia coli during Initial Contact with Cattle Colonic Explants. Microorganisms 2020; 8:E1662. [PMID: 33120988 PMCID: PMC7693793 DOI: 10.3390/microorganisms8111662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 01/10/2023] Open
Abstract
Foodborne pathogens are a public health threat globally. Shiga toxin-producing Escherichia coli (STEC), particularly O26, O111, and O157 STEC, are often associated with foodborne illness in humans. To create effective preharvest interventions, it is critical to understand which factors STEC strains use to colonize the gastrointestinal tract of cattle, which serves as the reservoir for these pathogens. Several colonization factors are known, but little is understood about initial STEC colonization factors. Our objective was to identify these factors via contrasting gene expression between nonpathogenic E. coli and STEC. Colonic explants were inoculated with nonpathogenic E. coli strain MG1655 or STEC strains (O26, O111, or O157), bacterial colonization levels were determined, and RNA was isolated and sequenced. STEC strains adhered to colonic explants at numerically but not significantly higher levels compared to MG1655. After incubation with colonic explants, flagellin (fliC) was upregulated (log2 fold-change = 4.0, p < 0.0001) in O157 STEC, and collectively, Lon protease (lon) was upregulated (log2 fold-change = 3.6, p = 0.0009) in STEC strains compared to MG1655. These results demonstrate that H7 flagellum and Lon protease may play roles in early colonization and could be potential targets to reduce colonization in cattle.
Collapse
Affiliation(s)
- Zachary R. Stromberg
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA;
| | - Rick E. Masonbrink
- Genome Informatics Facility, Iowa State University, Ames, IA 50011, USA;
| | - Melha Mellata
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA;
| |
Collapse
|
13
|
Menge C. The Role of Escherichia coli Shiga Toxins in STEC Colonization of Cattle. Toxins (Basel) 2020; 12:toxins12090607. [PMID: 32967277 PMCID: PMC7551371 DOI: 10.3390/toxins12090607] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 11/20/2022] Open
Abstract
Many cattle are persistently colonized with Shiga toxin-producing Escherichia coli (STEC) and represent a major source of human infections with human-pathogenic STEC strains (syn. enterohemorrhagic E. coli (EHEC)). Intervention strategies most effectively protecting humans best aim at the limitation of bovine STEC shedding. Mechanisms enabling STEC to persist in cattle are only partialy understood. Cattle were long believed to resist the detrimental effects of Shiga toxins (Stxs), potent cytotoxins acting as principal virulence factors in the pathogenesis of human EHEC-associated diseases. However, work by different groups, summarized in this review, has provided substantial evidence that different types of target cells for Stxs exist in cattle. Peripheral and intestinal lymphocytes express the Stx receptor globotriaosylceramide (Gb3syn. CD77) in vitro and in vivo in an activation-dependent fashion with Stx-binding isoforms expressed predominantly at early stages of the activation process. Subpopulations of colonic epithelial cells and macrophage-like cells, residing in the bovine mucosa in proximity to STEC colonies, are also targeted by Stxs. STEC-inoculated calves are depressed in mounting appropriate cellular immune responses which can be overcome by vaccination of the animals against Stxs early in life before encountering STEC. Considering Stx target cells and the resulting effects of Stxs in cattle, which significantly differ from effects implicated in human disease, may open promising opportunities to improve existing yet insufficient measures to limit STEC carriage and shedding by the principal reservoir host.
Collapse
Affiliation(s)
- Christian Menge
- Friedrich-Loeffler-Institut/Federal Research Institute for Animal Health, Institute of Molecular Pathogenesis, D-07743 Jena, Germany
| |
Collapse
|
14
|
Dey DK, Kang SC. Weissella confusa DD_A7 pre-treatment to zebrafish larvae ameliorates the inflammation response against Escherichia coli O157:H7. Microbiol Res 2020; 237:126489. [PMID: 32464536 DOI: 10.1016/j.micres.2020.126489] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/14/2020] [Accepted: 04/25/2020] [Indexed: 11/17/2022]
Abstract
Increasing multidrug-resistant pathogenic bacterial contamination in the environment has become the leading cause of food poisoning, resulting in life-threatening conditions due to late detection and limited therapeutic options. Escherichia coli O157:H7 is one such pathogen which is severely affecting the environmental livestock and ultimately leads to human infection. In this context, probiotics could be a useful strategy to minimize the growth of pathogens, as they produce several antimicrobial compounds and shows an exclusive competitive behavior against the pathogens. Therefore, supplementation of probiotics is wieldy accepted in the field of agriculture for the maintenance of animal's health. Previously, we reported that W. confusa DD_A7 possesses anti-bacterial and immune-stimulatory activity in-vitro. Therefore, in the present study, we investigated the impact of oral-administration of DD_A7 powder against E. coli O157:H7. The 6 days post-fertilized zebrafish larvae were used to evaluate the pathogenicity of the microbe. 1 × 108 CFU/ml of E. coli O157:H7 effectively induced the inflammatory response in zebrafish larvae. Where 1 × 108 CFU/ml DD_A7 pre-treatment prolonged the survivability of zebrafish larvae and improved the immune response of zebrafish larvae against pathogenic infection. The antibacterial property of DD_A7 against the pathogen correlated with the significant reduction of oxidative stress and host inflammatory response, by inhibiting NF-κB and its downstream signaling pathway. The findings demonstrated the prophylactic activity of DD_A7 suggesting that its supplementation improved the host defense mechanism by reducing oxidative stress. The growth of pathogen was effectively suppressed in the DD_A7 pre-treated larvae and maintained a healthy gastrointestinal environment in the zebrafish model.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Department of Biotechnology, Daegu University, Jillyang, Naeri-ri, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| | - Sun Chul Kang
- Department of Biotechnology, College of Engineering, Daegu University, Jillyang, Naeri-ri, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
15
|
Josenhans C, Müthing J, Elling L, Bartfeld S, Schmidt H. How bacterial pathogens of the gastrointestinal tract use the mucosal glyco-code to harness mucus and microbiota: New ways to study an ancient bag of tricks. Int J Med Microbiol 2020; 310:151392. [DOI: 10.1016/j.ijmm.2020.151392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
|
16
|
Li S, Peppelenbosch MP, Smits R. Bacterial biofilms as a potential contributor to mucinous colorectal cancer formation. Biochim Biophys Acta Rev Cancer 2019; 1872:74-79. [DOI: 10.1016/j.bbcan.2019.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023]
|
17
|
Xue Y, Du M, Zhu MJ. Raspberry extract prevents NLRP3 inflammasome activation in gut epithelial cells induced by pathogenic Escherichia coli. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
18
|
Wang G, Zhang Y, Song X, Xia Y, Lai PFH, Ai L. Lactobacillus caseiLC2W can inhibit the colonization ofEscherichia coliO157:H7in vivoand reduce the severity of colitis. Food Funct 2019; 10:5843-5852. [DOI: 10.1039/c9fo01390c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
L. caseiLC2W can inhibit the colonization of O157:H7 in mice and its prevention effect is stronger than the treatment effect.
Collapse
Affiliation(s)
- Guangqiang Wang
- Shanghai Engineering Research Center of Food Microbiology
- School of Medical Instrument and Food Engineering
- University of Shanghai for Science and Technology
- Shanghai 200093
- China
| | - Ying Zhang
- Shanghai Engineering Research Center of Food Microbiology
- School of Medical Instrument and Food Engineering
- University of Shanghai for Science and Technology
- Shanghai 200093
- China
| | - Xin Song
- Shanghai Engineering Research Center of Food Microbiology
- School of Medical Instrument and Food Engineering
- University of Shanghai for Science and Technology
- Shanghai 200093
- China
| | - Yongjun Xia
- Shanghai Engineering Research Center of Food Microbiology
- School of Medical Instrument and Food Engineering
- University of Shanghai for Science and Technology
- Shanghai 200093
- China
| | - Phoency F.-H. Lai
- Shanghai Engineering Research Center of Food Microbiology
- School of Medical Instrument and Food Engineering
- University of Shanghai for Science and Technology
- Shanghai 200093
- China
| | - Lianzhong Ai
- Shanghai Engineering Research Center of Food Microbiology
- School of Medical Instrument and Food Engineering
- University of Shanghai for Science and Technology
- Shanghai 200093
- China
| |
Collapse
|
19
|
Shekar A, Ramlal S, Jeyabalaji JK, Sripathy MH. Intranasal immunization of cocktail/fusion protein containing Tir along with ΔG active fragment of Zot as mucosal adjuvant confers enhanced immunogenicity and reduces E. coli O157:H7 shedding in mice. Int Immunopharmacol 2018; 63:211-219. [PMID: 30103196 DOI: 10.1016/j.intimp.2018.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/19/2018] [Accepted: 08/02/2018] [Indexed: 11/17/2022]
Abstract
Ruminants are the major reservoirs of Escherichia coli O157:H7 and its fecal shedding mainly act as a source of entry of this pathogen into the human food chain. In humans, E. coli O157:H7 infection causes diarrhea, hemorrhagic colitis and hemolytic uremic syndrome. Intimate adherence of E. coli O157:H7 is mediated by Translocated intimin receptor (Tir) to which intimin binds in the host cell. Since E. coli O157:H7 colonizes intestinal epithelium, the mucosal vaccine has a potential to prevent its colonization. Zonula occludens toxin (Zot) of Vibrio cholerae transiently, reversibly alters epithelial tight junction structure to increase mucosal permeability of macromolecules via paracellular route. The C-terminal region of Zot (ΔG) responsible for this function could be used for mucosal antigen delivery. Therefore, we employed individual (Tir), cocktail (ΔG + Tir), fusion protein (ΔG-Tir) and assessed the efficacy of its intranasal immunization on immunogenicity and fecal shedding of E. coli O157:H7 in streptomycin treated mouse model. Compared to control, ΔG + Tir, ΔG-Tir immunized mice elicited significant antigen specific antibody titers in serum (IgG, IgA) and feces (IgA), whereas Tir immunized mice induced only serum IgG titer. Cytokine analysis revealed mixed Th1/Th2 type immune response in case of ΔG + Tir, ΔG-Tir group while that of Tir group was solely Th2 type. Tir, ΔG + Tir and ΔG-Tir immunized mice showed reduction in shedding of E. coli O157:H7 compared to control group. However, ΔG-Tir immunized group performed better than ΔG + Tir, Tir group in reducing fecal shedding. Overall, our results demonstrate that intranasal immunization of ΔG-Tir induces effective systemic, mucosal, cellular immune responses and represents a promising mucosal subunit vaccine to prevent E. coli O157:H7 colonization.
Collapse
|
20
|
Affiliation(s)
- Christopher C L Chase
- Department of Veterinary and Biomedical Sciences, South Dakota State University, PO Box 2175, SAR Room 125, North Campus Drive, Brookings, SD 57007, USA.
| |
Collapse
|
21
|
Wang S, Cao Y, Deng S, Jiang X, Wang J, Zhang X, Zhang J, Liu G, Lian Z. Overexpression of Toll-like Receptor 4-linked Mitogen-activated Protein Kinase Signaling Contributes to Internalization of Escherichia coli in Sheep. Int J Biol Sci 2018; 14:1022-1032. [PMID: 29989103 PMCID: PMC6036738 DOI: 10.7150/ijbs.25275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/28/2018] [Indexed: 12/17/2022] Open
Abstract
Escherichia coli is one of the most common causal pathogens of mastitis in milk-producing mammals. Toll-like receptor 4 (TLR4) is important for host recognition of this bacteria. Increased activation of TLR4 can markedly enhance the internalization of E. coli. In this study, the relationship between TLR4 and mitogen-activated protein kinase (MAPK) signaling pathways in mediating E. coli internalization was evaluated in sheep monocytes. Using a TLR4-overexpressing transgenic (Tg) sheep model, we explored the bacterial internalization mechanism in sheep. We found that monocytes of Tg sheep could phagocytize more bacteria and exhibited higher adhesive capacity. The specific inhibition of p38 MAPK or c-Jun N-terminal kinase (JNK) or extracellular signal-regulated kinases (ERKs) reduced TLR4-dependent internalization of bacteria into sheep monocytes. Furthermore, the inhibition of MAPK signaling down-regulated the adhesive capacity of monocytes and the expression of scavenger receptors and adhesion molecules. Taken together, the overexpression of TLR4 in transgenic sheep enhanced the internalization of E. coli via MAPK signaling.
Collapse
Affiliation(s)
- Sutian Wang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yang Cao
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shoulong Deng
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaojing Jiang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiahao Wang
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | | | - Jinlong Zhang
- Tianjin Institute of Animal Sciences, Tianjin, China
| | - Guoshi Liu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhengxing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
22
|
Allaire JM, Morampudi V, Crowley SM, Stahl M, Yu H, Bhullar K, Knodler LA, Bressler B, Jacobson K, Vallance BA. Frontline defenders: goblet cell mediators dictate host-microbe interactions in the intestinal tract during health and disease. Am J Physiol Gastrointest Liver Physiol 2018; 314:G360-G377. [PMID: 29122749 PMCID: PMC5899238 DOI: 10.1152/ajpgi.00181.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Goblet cells (GCs) are the predominant secretory epithelial cells lining the luminal surface of the mammalian gastrointestinal (GI) tract. Best known for their apical release of mucin 2 (Muc2), which is critical for the formation of the intestinal mucus barrier, GCs have often been overlooked for their active contributions to intestinal protection and host defense. In part, this oversight reflects the limited tools available to study their function but also because GCs have long been viewed as relatively passive players in promoting intestinal homeostasis and host defense. In light of recent studies, this perspective has shifted, as current evidence suggests that Muc2 as well as other GC mediators are actively released into the lumen to defend the host when the GI tract is challenged by noxious stimuli. The ability of GCs to sense and respond to danger signals, such as bacterial pathogens, has recently been linked to inflammasome signaling, potentially intrinsic to the GCs themselves. Moreover, further work suggests that GCs release Muc2, as well as other mediators, to modulate the composition of the gut microbiome, leading to both the expansion as well as the depletion of specific gut microbes. This review will focus on the mechanisms by which GCs actively defend the host from noxious stimuli, as well as describe advanced technologies and new approaches by which their responses can be addressed. Taken together, we will highlight current insights into this understudied, yet critical, aspect of intestinal mucosal protection and its role in promoting gut defense and homeostasis.
Collapse
Affiliation(s)
- Joannie M. Allaire
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vijay Morampudi
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shauna M. Crowley
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Stahl
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hongbing Yu
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kirandeep Bhullar
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leigh A. Knodler
- 2Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Brian Bressler
- 3Division of Gastroenterology, Department of Medicine, St. Paul’s Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevan Jacobson
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce A. Vallance
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
23
|
Suppressing autophagy: a strategy by Escherichia coli O157:H7 for its survival on host epithelial cells. Cell Death Dis 2018; 9:64. [PMID: 29352117 PMCID: PMC5833748 DOI: 10.1038/s41419-017-0095-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 11/09/2022]
|
24
|
Escherichia coli O157:H7 suppresses host autophagy and promotes epithelial adhesion via Tir-mediated and cAMP-independent activation of protein kinase A. Cell Death Discov 2017; 3:17055. [PMID: 28975041 PMCID: PMC5624281 DOI: 10.1038/cddiscovery.2017.55] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 07/07/2017] [Indexed: 11/18/2022] Open
Abstract
Autophagy is a pivotal innate immune response that not only degrades cytosolic components, but also serves as one of the critical antimicrobial mechanisms eliminating intracellular pathogens. However, its role in host defense against extracellular pathogens is largely unknown. Here we showed that E. coli O157:H7 altered autophagy to evade host defense and facilitate adhesion. Enhancing host cell autophagy with tumor necrosis factor (TNF), host starvation or rapamycin reduced the adherence of E. coli O157:H7 to HT-29 cells. As a key regulator of autophagy, protein kinase A (PKA) was activated by E. coli O157:H7 infection. PKA inhibition by H89 abrogated E. coli O157:H7 inhibition of autophagy and prevented bacterial epithelial adhesion. Thus, PKA had a mediatory role in blocking autophagy and E. coli O157:H7 epithelial adhesion. Furthermore, deletion of translocated intimin receptor (tir) prevented PKA activation, whereas ectopic tir expression in a Δtir mutant strain restored its ability to activate PKA and inhibited autophagy in host cells. This indicated that Tir and PKA played pivotal roles in manipulating host autophagy during infection. Consistent with autophagy inhibition, E. coli O157:H7 infection inhibited endoplasmic reticulum (ER) stress in HT-29 cells, which was reversed by TNF, starvation, or H89 treatment. Additionally, E. coli O157:H7-induced PKA activation suppressed extracellular signal-regulated kinase 1/2 (ERK1/2) activation and enhanced phosphatidylinositol 3-kinase/Akt (PI3K/Akt) signaling, thereby repressing autophagic signaling. Conversely, PKA inhibition prevented downregulation of ERK1/2 signaling due to E. coli O157:H7 infection. In summary, E. coli O157:H7 inhibited host autophagy via Tir-mediated PKA activation that favored bacterial persistence on intestinal epithelial cell surfaces.
Collapse
|
25
|
Sicard JF, Le Bihan G, Vogeleer P, Jacques M, Harel J. Interactions of Intestinal Bacteria with Components of the Intestinal Mucus. Front Cell Infect Microbiol 2017; 7:387. [PMID: 28929087 PMCID: PMC5591952 DOI: 10.3389/fcimb.2017.00387] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022] Open
Abstract
The human gut is colonized by a variety of large amounts of microbes that are collectively called intestinal microbiota. Most of these microbial residents will grow within the mucus layer that overlies the gut epithelium and will act as the first line of defense against both commensal and invading microbes. This mucus is essentially formed by mucins, a family of highly glycosylated protein that are secreted by specialize cells in the gut. In this Review, we examine how commensal members of the microbiota and pathogenic bacteria use mucus to their advantage to promote their growth, develop biofilms and colonize the intestine. We also discuss how mucus-derived components act as nutrient and chemical cues for adaptation and pathogenesis of bacteria and how bacteria can influence the composition of the mucus layer.
Collapse
Affiliation(s)
- Jean-Félix Sicard
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Guillaume Le Bihan
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Philippe Vogeleer
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Mario Jacques
- Regroupement de Recherche Pour un Lait de Qualité Optimale (Op+Lait), Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| | - Josée Harel
- Centre de Recherche en Infectiologie Porcine et Aviaire, Faculté de Médecine Vétérinaire, Université de MontréalSaint-Hyacinthe, QC, Canada
| |
Collapse
|
26
|
Functional Enterospheres Derived In Vitro from Human Pluripotent Stem Cells. Stem Cell Reports 2017; 9:897-912. [PMID: 28867347 PMCID: PMC5599260 DOI: 10.1016/j.stemcr.2017.07.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023] Open
Abstract
Intestinal organoids derived from human pluripotent stem cells (hPSCs) are valuable in vitro research models that enable simplified access to human gastrointestinal tissues. Here, we report the in vitro generation of enterospheres (hEnS) from hPSC-derived gastrointestinal epithelial precursors. hEnS are cystic spheroids with a simple uniform structure composed entirely of intestinal epithelium. hEnS express markers of mature brush border cells and share a transcriptome profile similar to that of more mature intestinal organoids. Modulation of signaling cues enables control of hEnS growth and differentiation, including long-term propagation. We show that hEnS can be exploited for functional studies: hEnS display an innate immune response when treated with enteric pathogens, and transgenic modification of hEnS with a fluorescence cell-cycle reporter enables hEnS-forming stem cell enrichment. Our work establishes hEnS as an accessible and tractable in vitro modeling system for studying human gastrointestinal biology.
Collapse
|
27
|
Xue Y, Du M, Zhu MJ. Quercetin suppresses NLRP3 inflammasome activation in epithelial cells triggered by Escherichia coli O157:H7. Free Radic Biol Med 2017; 108:760-769. [PMID: 28476502 DOI: 10.1016/j.freeradbiomed.2017.05.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/22/2017] [Accepted: 05/01/2017] [Indexed: 12/28/2022]
Abstract
Inflammatory responses elicited by LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is induced by a wide variety of stress signals including infectious agents and cellular disorders. E. coli O157:H7 causes serious gastrointestinal diseases that results in severe inflammation and oxidative stress, causing host cell damage. In this study, we found that E. coli O157:H7 infection induced NLRP3 assembly, caspase-1 activation and interleukin (IL)-1β and IL-18 release in Caco-2 cells. Infection also resulted in mitochondrial dysfunction with disrupted mitochondrial potential and mitochondrial complex-I activity, as well as the cytosolic release of cytochrome c and altered mitochondrial respiratory chain. The damage of mitochondria led to increased production of reactive oxygen species (ROS) and cytosolic release of mitochondrial DNA. Moreover, ROS was required for E. coli O157:H7 induced NLRP3 assembly as inhibiting mitochondrial ROS release by ROS scavengers Mito-TEMPO and N-acetylcysteine abrogated NLRP3 inflammasome activation in Caco-2 cells in response to E. coli O157:H7. Quercetin, one of the most important flavonoids in plant origin foods, had a protective role in inhibiting NLRP3 activation upon E. coli O157:H7 infection by protecting mitochondrial integrity and inhibiting mitochondrial ROS release. In addition, E. coli O157:H7 infection inhibited the host autophagy while quercetin treatment augmented autophagy activation, which further blocked ROS generation and IL-1β and IL-18 release. In summary, E. coli O157:H7 infection induced mitochondrial ROS release and NLRP3 assembly in host cells, while quercetin exerted a preventive role in host cells upon E. coli O157:H7 infection partially due to prevention of ROS production and activation of autophagy.
Collapse
Affiliation(s)
- Yansong Xue
- School of Food Science, Washington State University, Pullman, WA 99164, USA
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, WA 99164, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
28
|
Complete Genome Sequence of Escherichia coli Strain M8, Isolated from ob/ob Mice. GENOME ANNOUNCEMENTS 2017; 5:5/22/e00449-17. [PMID: 28572322 PMCID: PMC5454205 DOI: 10.1128/genomea.00449-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Escherichia coli is one of the common inhabitants of the mammalian gastrointestinal track. We isolated a strain from an ob/ob mouse and performed whole-genome sequencing, which yielded a chromosome of ~5.1 Mb and three plasmids of ~160 kb, ~6 kb, and ~4 kb.
Collapse
|
29
|
How do they stick together? Bacterial adhesins implicated in the binding of bacteria to the human gastrointestinal mucins. Biochem Soc Trans 2017; 45:389-399. [PMID: 28408479 DOI: 10.1042/bst20160167] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/16/2016] [Accepted: 01/23/2017] [Indexed: 01/04/2023]
Abstract
The gastrointestinal mucosal surface is the primary interface between internal host tissues and the vast microbiota. Mucins, key components of mucus, are high-molecular-weight glycoproteins characterized by the presence of many O-linked oligosaccharides to the core polypeptide. They play many biological functions, helping to maintain cellular homeostasis and to establish symbiotic relationships with complex microbiota. Mucin O-glycans exhibit a huge variety of peripheral sequences implicated in the binding of bacteria to the mucosal tissues, thereby playing a key role in the selection of specific species and in the tissue tropism displayed by commensal and pathogenic bacteria. Bacteria have evolved numerous strategies to colonize host mucosae, and among these are modulation of expression of cell surface adhesins which allow bacteria to bind to mucins. However, despite well structurally characterized adhesins and lectins, information on the nature and structure of oligosaccharides recognized by bacteria is still disparate. This review summarizes the current knowledge on the structure of epithelial mucin O-glycans and the interaction between host and commensal or pathogenic bacteria mediated by mucins.
Collapse
|
30
|
Hews CL, Tran SL, Wegmann U, Brett B, Walsham ADS, Kavanaugh D, Ward NJ, Juge N, Schüller S. The StcE metalloprotease of enterohaemorrhagic Escherichia coli reduces the inner mucus layer and promotes adherence to human colonic epithelium ex vivo. Cell Microbiol 2017; 19. [PMID: 28054754 PMCID: PMC5434857 DOI: 10.1111/cmi.12717] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 12/20/2016] [Accepted: 12/30/2016] [Indexed: 12/18/2022]
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) is a major foodborne pathogen and tightly adheres to human colonic epithelium by forming attaching/effacing lesions. To reach the epithelial surface, EHEC must penetrate the thick mucus layer protecting the colonic epithelium. In this study, we investigated how EHEC interacts with the intestinal mucus layer using mucin‐producing LS174T colon carcinoma cells and human colonic mucosal biopsies. The level of EHEC binding and attaching/effacing lesion formation in LS174T cells was higher compared to mucin‐deficient colon carcinoma cell lines, and initial adherence was independent of the presence of flagellin, Escherichia coli common pilus, or long polar fimbriae. Although EHEC infection did not affect gene expression of secreted mucins, it resulted in reduced MUC2 glycoprotein levels. This effect was dependent on the catalytic activity of the secreted metalloprotease StcE, which reduced the inner mucus layer and thereby promoted EHEC access and binding to the epithelium in vitro and ex vivo. Given the lack of efficient therapies against EHEC infection, StcE may represent a suitable target for future treatment and prevention strategies.
Collapse
Affiliation(s)
- Claire L Hews
- Gut Health and Food Safety Programme, Institute of Food Research, Norwich, UK.,School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Seav-Ly Tran
- Norwich Medical School, University of East Anglia, Norwich, UK.,Gut Health and Food Safety Programme, Institute of Food Research, Norwich, UK
| | - Udo Wegmann
- Gut Health and Food Safety Programme, Institute of Food Research, Norwich, UK
| | - Bernard Brett
- Department of Gastroenterology, Norfolk and Norwich University Hospital, Norwich, UK.,Department of Gastroenterology, James Paget University Hospital, Great Yarmouth, UK
| | - Alistair D S Walsham
- Norwich Medical School, University of East Anglia, Norwich, UK.,Gut Health and Food Safety Programme, Institute of Food Research, Norwich, UK
| | - Devon Kavanaugh
- Gut Health and Food Safety Programme, Institute of Food Research, Norwich, UK
| | - Nicole J Ward
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Nathalie Juge
- Gut Health and Food Safety Programme, Institute of Food Research, Norwich, UK
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich, UK.,Gut Health and Food Safety Programme, Institute of Food Research, Norwich, UK
| |
Collapse
|
31
|
Xue Y, Zhang S, Du M, Zhu MJ. Dandelion extract suppresses reactive oxidative species and inflammasome in intestinal epithelial cells. J Funct Foods 2017. [DOI: 10.1016/j.jff.2016.11.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
32
|
Sanchez-Villamil J, Tapia-Pastrana G, Navarro-Garcia F. Pathogenic Lifestyles of E. coli Pathotypes in a Standardized Epithelial Cell Model Influence Inflammatory Signaling Pathways and Cytokines Secretion. Front Cell Infect Microbiol 2016; 6:120. [PMID: 27774437 PMCID: PMC5054702 DOI: 10.3389/fcimb.2016.00120] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/21/2016] [Indexed: 12/21/2022] Open
Abstract
Inflammatory response is key for the host defense against diarrheagenic Escherichia coli and contributes to the pathogenesis of the disease but there is not a comparative study among different diarrheagenic pathotypes. We analyzed the inflammatory response induced by five diarrheagenic pathotypes in a HT-29 cell infection model. The model was unified to reproduce the pathogenesis of each pathotype. To compare the inflammatory responses we evaluated: (i) nuclear NF-κB and ERK1/2 translocation by confocal microscopy; (ii) kinetics of activation by each pathway detecting p65 and ERK1/2 phosphorylation by Western blotting; (iii) pathways modulation through bacterial infections with or without co-stimulation with TNF-α or EGF; (iv) cytokine profile induced by each pathotype with and without inhibitors of each pathway. EHEC but mainly EPEC inhibited translocation and activation of p65 and ERK1/2 pathways, as well as cytokines secretion; inhibition of p65 and ERK1/2 phosphorylation prevailed in the presence of TNF-α and EGF, respectively. Intracellular strains, EIEC/Shigella flexneri, caused a strong translocation, activation, and cytokines secretion but they could not inhibit TNF-α and EGF stimulation. ETEC and mainly EAEC caused a moderate translocation, but a differential activation, and high cytokines secretion; interestingly TNF-α and EGF stimulation did no modify p65 and ERK1/2 activation. The use of inhibitors of NF-κB and/or ERK1/2 showed that NF-κB is crucial for cytokine induction by the different pathotypes; only partially depended on ERK1/2 activation. Thus, in spite of their differences, the pathotypes can also be divided in three groups according to their inflammatory response as those (i) that inject effectors to cause A/E lesion, which are able to inhibit NF-κB and ERK1/2 pathways, and cytokine secretion; (ii) with fimbrial adherence and toxin secretion with a moderate inhibition of both pathways but high cytokines secretion through autocrine cytokine regulation; and (iii) the intracellular bacteria that induce the highest pathways activation and cytokines secretion by using different activation mechanisms. This study provides a comprehensive analysis of how the different pathogenesis schemes of E. coli pathotypes manipulate inflammatory signaling pathways, which leads to a specific proinflammatory cytokine secretion in a cell model infection that reproduce the hallmarks of infection of each pathotype.
Collapse
Affiliation(s)
- Javier Sanchez-Villamil
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, Mexico
| | - Gabriela Tapia-Pastrana
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, Mexico
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional México City, Mexico
| |
Collapse
|
33
|
Loktionov A, Chhaya V, Bandaletova T, Poullis A. Assessment of cytology and mucin 2 in colorectal mucus collected from patients with inflammatory bowel disease: Results of a pilot trial. J Gastroenterol Hepatol 2016; 31:326-33. [PMID: 26248500 DOI: 10.1111/jgh.13083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/06/2015] [Accepted: 07/08/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Non-invasive diagnosis of colorectal disease remains problematic, fecal biomarkers presenting the only current option. Colorectal mucus is the diagnostically informative element of stool samples, but its separation from stool is difficult. We aimed to: (i) test a novel method of non-invasive colorectal mucus sampling in a pilot clinical trial; (ii) evaluate sampling method acceptance by study participants; (iii) characterize the collected material cytologically; and (iv) assess feasibility of quantitative protein analysis in the samples. METHODS A total of 141 patients with IBD (58), IBS (50), and healthy controls (33) participated in the study. Samples rich in colorectal mucus were self-collected by swabbing the anal area immediately following defecation. Collected samples were examined cytologically and subjected to quantitative analysis for total protein and mucin 2 (MUC2). RESULTS The novel sampling technique was assessed as "good" or "adequate" by 96% of study participants. A total of 55% of the collected samples were free of fecal contamination. Cytology showed large numbers of well preserved inflammatory cells in IBD cases. Total protein values varied in all groups, being affected by fecal contamination. MUC2 levels were similar among all IBD-free individuals (control and IBS groups) and elevated in IBD patients (p < 0.001). MUC2 measurement applied as a test for IBD detection provided sensitivity = 72.4% and specificity = 86.7%. CONCLUSIONS A novel non-invasive method for collecting human colorectal mucus has been successfully tested. The method was very well accepted by trial participants. The results have proven high quality of collected samples for both cytological investigation and diagnostic biomarker analysis.
Collapse
Affiliation(s)
| | - Vivek Chhaya
- Department of Gastroenterology, St George's Hospital, London, UK
| | | | - Andrew Poullis
- Department of Gastroenterology, St George's Hospital, London, UK
| |
Collapse
|
34
|
Biomarkers of Gastrointestinal Host Responses to Microbial Infections. Mol Microbiol 2016. [DOI: 10.1128/9781555819071.ch46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
35
|
Heat-labile enterotoxin of Escherichia coli promotes intestinal colonization of Salmonella enterica. Comp Immunol Microbiol Infect Dis 2015; 43:1-7. [PMID: 26616654 DOI: 10.1016/j.cimid.2015.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/31/2015] [Accepted: 09/12/2015] [Indexed: 02/04/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important cause of infantile and travellers' diarrhoea, which poses a serious health burden, especially in developing countries. In addition, ETEC bacteria are a major cause of illness and death in neonatal and recently weaned pigs. The production of a heat-labile enterotoxin (LT) promotes the colonization and pathogenicity of ETEC and may exacerbate co-infections with other enteric pathogens such as Salmonella enterica. We showed that the intraintestinal presence of LT dramatically increased the intestinal Salmonella Typhimurium load in experimentally inoculated pigs. This could not be explained by direct alteration of the invasion or survival capacity of Salmonella in enterocytes, in vitro. However, we demonstrated that LT affects the enteric mucus layer composition in a mucus-secreting goblet cell line by significantly decreasing the expression of mucin 4. The current results show that LT alters the intestinal mucus composition and aggravates a Salmonella Typhimurium infection, which may result in the exacerbation of the diarrhoeal illness.
Collapse
|
36
|
Escherichia coli LF82 differentially regulates ROS production and mucin expression in intestinal epithelial T84 cells: implication of NOX1. Inflamm Bowel Dis 2015; 21:1018-26. [PMID: 25822013 DOI: 10.1097/mib.0000000000000365] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Increased reactive oxygen species (ROS) production is associated with inflamed ileal lesions in Crohn's disease colonized by pathogenic adherent-invasive Escherichia coli LF82. We investigated whether such ileal bacteria can modulate ROS production by epithelial cells, thus impacting on inflammation and mucin expression. METHODS Ileal bacteria from patients with Crohn's disease were incubated with cultured epithelial T84 cells, and ROS production was assayed using the luminol-amplified chemiluminescence method. The gentamicin protection assay was used for bacterial invasion of T84 cell. The expression of NADPH oxidase (NOX) subunits, mucin, and IL-8 was analyzed by quantitative real-time PCR and Western blots. Involvement of NOX and ROS was analyzed using diphenyleneiodonium (DPI) and N-acetylcysteine (NAC). RESULTS Among different bacteria tested, only LF82 induced an increase of ROS production by T84 cells in a dose-dependent manner. This response was inhibited by DPI and NAC. Heat- or ethanol-attenuated LF82 bacteria and the mutant LF82ΔFimA, which does not express pili type 1 and poorly adheres to epithelial cells, did not induce the oxidative response. The LF82-induced oxidative response coincides with its invasion in T84 cells, and both processes were inhibited by DPI. Also, we observed an increased expression of NOX1 and NOXO1 in response to LF82 bacteria versus the mutant LF82ΔFimA. Furthermore, LF82 inhibited mucin gene expression (MUC2 and MUC5AC) in T84 cells while increasing the chemotactic IL-8 expression, both in a DPI-sensitive manner. CONCLUSIONS Adherent-invasive E. coli LF82 induced ROS production by intestinal NADPH oxidase and altered mucin and IL-8 expression, leading to perpetuation of inflammatory lesions in Crohn's disease.
Collapse
|
37
|
Strain-dependent cellular immune responses in cattle following Escherichia coli O157:H7 colonization. Infect Immun 2014; 82:5117-31. [PMID: 25267838 DOI: 10.1128/iai.02462-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 causes hemorrhagic diarrhea and potentially fatal renal failure in humans. Ruminants are considered to be the primary reservoir for human infection. Vaccines that reduce shedding in cattle are only partially protective, and their underlying protective mechanisms are unknown. Studies investigating the response of cattle to colonization generally focus on humoral immunity, leaving the role of cellular immunity unclear. To inform future vaccine development, we studied the cellular immune responses of cattle during EHEC O157:H7 colonization. Calves were challenged either with a phage type 21/28 (PT21/28) strain possessing the Shiga toxin 2a (Stx2a) and Stx2c genes or with a PT32 strain possessing the Stx2c gene only. T-helper cell-associated transcripts at the terminal rectum were analyzed by reverse transcription-quantitative PCR (RT-qPCR). Induction of gamma interferon (IFN-γ) and T-bet was observed with peak expression of both genes at 7 days in PT32-challenged calves, while upregulation was delayed, peaking at 21 days, in PT21/28-challenged calves. Cells isolated from gastrointestinal lymph nodes demonstrated antigen-specific proliferation and IFN-γ release in response to type III secreted proteins (T3SPs); however, responsiveness was suppressed in cells isolated from PT32-challenged calves. Lymph node cells showed increased expression of the proliferation marker Ki67 in CD4(+) T cells from PT21/28-challenged calves, NK cells from PT32-challenged calves, and CD8(+) and γδ T cells from both PT21/28- and PT32-challenged calves following ex vivo restimulation with T3SPs. This study demonstrates that cattle mount cellular immune responses during colonization with EHEC O157:H7, the temporality of which is strain dependent, with further evidence of strain-specific immunomodulation.
Collapse
|