1
|
Kilari G, Tran J, Blyth GAD, Cobo ER. Human cathelicidin LL-37 rapidly disrupted colonic epithelial integrity. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184410. [PMID: 39837472 DOI: 10.1016/j.bbamem.2025.184410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
The intestinal barrier, held together by epithelial cells and intercellular tight junction (TJ) proteins, prevents the penetration of microbial pathogens. Concurrently, intestinal epithelial cells secrete antimicrobial peptides, including cathelicidin. Cathelicidin has direct antibacterial and immunomodulatory functions, although its role in intestinal integrity remains elusive. In this study, we demonstrate that direct stimulation of human colonic epithelial (T84) cells with human cathelicidin, LL-37, resulted in a rapid and transient increase in epithelial cell permeability. This increased permeability was associated with the TJ proteins occludin and claudin-2 degradation, mediated by these specific proteins' endocytosis and lysosomal degradation. While murine cathelicidin (CRAMP) failed to modify T84 cell permeability, LL-37 degraded TJ proteins in murine rectal epithelial (CMT-93) cells. The stimulus of (CMT-93) cells with LL-37 aggravated the cell permeability and furthered TJ degradation provoked by the intestinal pathogen, attaching/effacing (A/E) Citrobacter rodentium (C. rodentium). The number of C. rodentium that colonized CMT-93 cells was not severely impacted by the presence of LL-37. While a temporary disruption of tight junctions by LL-37 may lead to a 'leaky gut,' this study demonstrates that LL-37 increases epithelial cell permeability by degrading TJ proteins occludin and claudin-2 through endocytosis and lysosomal degradation. These immunomodulatory actions occurring at concentrations lower than those microbicidal uncover a new guise for cathelicidin modulating the epithelial barrier against A/E pathogens. Recognizing native cathelicidin's functions in a specified disease setting (e.g., colitis) will help establish it as an anti-infectious immunomodulator.
Collapse
Affiliation(s)
- Geeta Kilari
- Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Jacquelyn Tran
- Faculty of Veterinary Medicine, University of Calgary, Canada
| | | | - Eduardo R Cobo
- Faculty of Veterinary Medicine, University of Calgary, Canada.
| |
Collapse
|
2
|
Huang Q, Zhang Y, Chu Q, Song H. The Influence of Polysaccharides on Lipid Metabolism: Insights from Gut Microbiota. Mol Nutr Food Res 2024; 68:e2300522. [PMID: 37933720 DOI: 10.1002/mnfr.202300522] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/06/2023] [Indexed: 11/08/2023]
Abstract
SCOPE Polysaccharides are complex molecules of more than ten monosaccharide residues interconnected through glycosidic linkages formed via condensation reactions. Polysaccharides are widely distributed in various food resources and have gained considerable attention due to their diverse biological activities. This review presented a critical analysis of the existing research literature on anti-obesity polysaccharides and investigates the complex interplay between their lipid-lowering activity and the gut microbiota, aiming to provide a comprehensive overview of the lipid-lowering properties of polysaccharides and the underlying mechanisms of action. METHODS AND RESULTS In this review, the study summarized the roles of polysaccharides in improving lipid metabolism via gut microbiota, including the remodeling of the intestinal barrier, reduction of inflammation, inhibition of pathogenic bacteria, reduction of trimethylamine N-oxide (TMAO) production, and regulation of the metabolism of short-chain fatty acids (SCFAs) and bile acids (BAs). CONCLUSION These mechanisms collectively contributed to the beneficial effects of polysaccharides on lipid metabolism and overall metabolic health. Furthermore, polysaccharide-based nanocarriers combined with gut microbiota have broad prospects for developing targeted and personalized therapies for hyperlipidemia and obesity.
Collapse
Affiliation(s)
- Qianqian Huang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, 210023, China
| | - Yanhui Zhang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, 210023, China
| | - Qiang Chu
- Tea Research Institute, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Haizhao Song
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, 210023, China
| |
Collapse
|
3
|
Srivastava P, Kim KS. Membrane Vesicles Derived from Gut Microbiota and Probiotics: Cutting-Edge Therapeutic Approaches for Multidrug-Resistant Superbugs Linked to Neurological Anomalies. Pharmaceutics 2022; 14:2370. [PMID: 36365188 PMCID: PMC9692612 DOI: 10.3390/pharmaceutics14112370] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Multidrug-resistant (MDR) superbugs can breach the blood-brain barrier (BBB), leading to a continuous barrage of pro-inflammatory modulators and induction of severe infection-related pathologies, including meningitis and brain abscess. Both broad-spectrum or species-specific antibiotics (β-lactamase inhibitors, polymyxins, vancomycin, meropenem, plazomicin, and sarecycline) and biocompatible poly (lactic-co-glycolic acid) (PLGA) nanoparticles have been used to treat these infections. However, new therapeutic platforms with a broad impact that do not exert off-target deleterious effects are needed. Membrane vesicles or extracellular vesicles (EVs) are lipid bilayer-enclosed particles with therapeutic potential owing to their ability to circumvent BBB constraints. Bacteria-derived EVs (bEVs) from gut microbiota are efficient transporters that can penetrate the central nervous system. In fact, bEVs can be remodeled via surface modification and CRISPR/Cas editing and, thus, represent a novel platform for conferring protection against infections breaching the BBB. Here, we discuss the latest scientific research related to gut microbiota- and probiotic-derived bEVs, and their therapeutic modifications, in terms of regulating neurotransmitters and inhibiting quorum sensing, for the treatment of neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases. We also emphasize the benefits of probiotic-derived bEVs to human health and propose a novel direction for the development of innovative heterologous expression systems to combat BBB-crossing pathogens.
Collapse
Affiliation(s)
| | - Kwang-sun Kim
- Department of Chemistry and Chemistry Institute of Functional Materials, Pusan National University, Busan 46241, Korea
| |
Collapse
|
4
|
Schwartz B, Vetvicka V. Review: β-glucans as Effective Antibiotic Alternatives in Poultry. Molecules 2021; 26:molecules26123560. [PMID: 34200882 PMCID: PMC8230556 DOI: 10.3390/molecules26123560] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/01/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
The occurrence of microbial challenges in commercial poultry farming causes significant economic losses. Antibiotics have been used to control diseases involving bacterial infection in poultry. As the incidence of antibiotic resistance turns out to be a serious problem, there is increased pressure on producers to reduce antibiotic use. With the reduced availability of antibiotics, poultry producers are looking for feed additives to stimulate the immune system of the chicken to resist microbial infection. Some β-glucans have been shown to improve gut health, to increase the flow of new immunocytes, increase macrophage function, stimulate phagocytosis, affect intestinal morphology, enhance goblet cell number and mucin-2 production, induce the increased expression of intestinal tight-junctions, and function as effective anti-inflammatory immunomodulators in poultry. As a result, β-glucans may provide a new tool for producers trying to reduce or eliminate the use of antibiotics in fowl diets. The specific activity of each β-glucan subtype still needs to be investigated. Upon knowledge, optimal β-glucan mixtures may be implemented in order to obtain optimal growth performance, exert anti-inflammatory and immunomodulatory activity, and optimized intestinal morphology and histology responses in poultry. This review provides an extensive overview of the current use of β glucans as additives and putative use as antibiotic alternative in poultry.
Collapse
Affiliation(s)
- Betty Schwartz
- Institute of Biochemistry, Food Science and Nutrition, The School of Nutritional Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 761001, Israel
- Correspondence:
| | - Vaclav Vetvicka
- Department of Pathology, University of Louisville, Louisville, KY 40202, USA;
| |
Collapse
|
5
|
Ledwaba SE, Costa DVS, Bolick DT, Giallourou N, Medeiros PHQS, Swann JR, Traore AN, Potgieter N, Nataro JP, Guerrant RL. Enteropathogenic Escherichia coli Infection Induces Diarrhea, Intestinal Damage, Metabolic Alterations, and Increased Intestinal Permeability in a Murine Model. Front Cell Infect Microbiol 2020; 10:595266. [PMID: 33392105 PMCID: PMC7773950 DOI: 10.3389/fcimb.2020.595266] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Enteropathogenic E. coli (EPEC) are recognized as one of the leading bacterial causes of infantile diarrhea worldwide. Weaned C57BL/6 mice pretreated with antibiotics were challenged orally with wild-type EPEC or escN mutant (lacking type 3 secretion system) to determine colonization, inflammatory responses and clinical outcomes during infection. Antibiotic disruption of intestinal microbiota enabled efficient colonization by wild-type EPEC resulting in growth impairment and diarrhea. Increase in inflammatory biomarkers, chemokines, cellular recruitment and pro-inflammatory cytokines were observed in intestinal tissues. Metabolomic changes were also observed in EPEC infected mice with changes in tricarboxylic acid (TCA) cycle intermediates, increased creatine excretion and shifts in gut microbial metabolite levels. In addition, by 7 days after infection, although weights were recovering, EPEC-infected mice had increased intestinal permeability and decreased colonic claudin-1 levels. The escN mutant colonized the mice with no weight loss or increased inflammatory biomarkers, showing the importance of the T3SS in EPEC virulence in this model. In conclusion, a murine infection model treated with antibiotics has been developed to mimic clinical outcomes seen in children with EPEC infection and to examine potential roles of selected virulence traits. This model can help in further understanding mechanisms involved in the pathogenesis of EPEC infections and potential outcomes and thus assist in the development of potential preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Solanka E. Ledwaba
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - Deiziane V. S. Costa
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - David T. Bolick
- Center for Global Health, Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Natasa Giallourou
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, London, England
| | | | - Jonathan R. Swann
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, London, England
| | - Afsatou N. Traore
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - Natasha Potgieter
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - James P. Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Richard L. Guerrant
- Center for Global Health, Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
6
|
Mucin-2 knockout is a model of intercellular junction defects, mitochondrial damage and ATP depletion in the intestinal epithelium. Sci Rep 2020; 10:21135. [PMID: 33273633 PMCID: PMC7713437 DOI: 10.1038/s41598-020-78141-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
The disruption of the protective intestinal barrier—the ‘leaky gut’—is a common complication of the inflammatory bowel disease. There is limited data on the mechanisms of the intestinal barrier disruption upon low-grade inflammation characteristic of patients with inflammatory bowel disease in clinical remission. Thus, animal models that recapitulate the complexity of chronic intestinal inflammation in vivo are of particular interest. In this study, we used Mucin-2 (Muc2) knockout mice predisposed to colitis to study intestinal barrier upon chronic inflammation. We used 4-kDa FITC-Dextran assay and transmission electron microscopy to demonstrate the increased intestinal permeability and morphological defects in intercellular junctions in Muc2 knockout mice. Confocal microscopy revealed the disruption of the apical F-actin cytoskeleton and delocalization of tight junction protein Claudin-3 from the membrane. We further demonstrate mitochondrial damage, impaired oxygen consumption and the reduction of the intestinal ATP content in Muc2 knockout mice. Finally, we show that chemically induced mitochondrial uncoupling in the wild type mice mimics the intestinal barrier disruption in vivo and causes partial loss of F-actin and membrane localization of Claudin-3. We propose that mitochondrial damage and metabolic shifts during chronic inflammation contribute to the leaky gut syndrome in Muc2 knockout animal model of colitis.
Collapse
|
7
|
Rashid F, Ahmad S, Irudayaraj JMK. Effect of Perfluorooctanoic Acid on the Epigenetic and Tight Junction Genes of the Mouse Intestine. TOXICS 2020; 8:toxics8030064. [PMID: 32872178 PMCID: PMC7560341 DOI: 10.3390/toxics8030064] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022]
Abstract
Perfluorooctanoic acid (PFOA) has been implicated in various toxicities including neurotoxicity, genotoxicity, nephrotoxicity, epigenetic toxicity, immunotoxicity, reproductive toxicity, and hepatotoxicity. However, information on the accumulation of PFOA in the intestine and its toxic effects on intestinal epigenetics and tight junction (TJ) genes is sparse. CD1 mice were dosed with PFOA (1, 5, 10, or 20 mg/kg/day) for 10 days, and its accumulation and induced alterations in the expression of epigenetic and tight junction genes in the small intestine and colon were evaluated using LC-MS and qPCR techniques. PFOA reduced the expression levels of DNA methyltransferases (Dnmt1, Dnmt3a, Dnmt3b) primarily in the small intestine whereas, in the colon, a decrease was observed only at high concentrations. Moreover, ten-eleven translocation genes (Tet2 and Tet3) expression was dysregulated in the small intestine, whereas in the colon Tets remained unaffected. The tight junction genes Claudins (Cldn), Occludin (Ocln), and Tight Junction Protein (Tjp) were also heavily altered in the small intestine. TJs responded differently across the gut, in proportion to PFOA dosing. Our study reveals that PFOA triggers DNA methylation changes and alters the expression of genes essential for maintaining the physical barrier of intestine, with more profound effects in the small intestine compared to the colon.
Collapse
Affiliation(s)
- Faizan Rashid
- Biomedical Research Center in Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA; (F.R.); (S.A.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Saeed Ahmad
- Biomedical Research Center in Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA; (F.R.); (S.A.)
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Joseph Maria Kumar Irudayaraj
- Biomedical Research Center in Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA; (F.R.); (S.A.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Correspondence: ; Tel.: +1-765-404-0499
| |
Collapse
|
8
|
Post-transcriptional air pollution oxidation to the cholesterol biosynthesis pathway promotes pulmonary stress phenotypes. Commun Biol 2020; 3:392. [PMID: 32699268 PMCID: PMC7376215 DOI: 10.1038/s42003-020-01118-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
The impact of environmentally-induced chemical changes in RNA has been fairly unexplored. Air pollution induces oxidative modifications such as 8-oxo-7,8-dihydroguanine (8-oxoG) in RNAs of lung cells, which could be associated with premature lung dysfunction. We develop a method for 8-oxoG profiling using immunocapturing and RNA sequencing. We find 42 oxidized transcripts in bronchial epithelial BEAS-2B cells exposed to two air pollution mixtures that recreate urban atmospheres. We show that the FDFT1 transcript in the cholesterol biosynthesis pathway is susceptible to air pollution-induced oxidation. This process leads to decreased transcript and protein expression of FDFT1, and reduced cholesterol synthesis in cells exposed to air pollution. Knockdown of FDFT1 replicates alterations seen in air pollution exposure such as transformed cell size and suppressed cytoskeleton organization. Our results argue of a possible novel biomarker and of an unseen mechanism by which air pollution selectively modifies key metabolic-related transcripts facilitating cell phenotypes in bronchial dysfunction. Gonzales-Rivera et al. develop a method for 8-oxoG profiling using immunocapturing and RNA sequencing. They show that the FDFT1 transcript is susceptible to air pollution-induced oxidation, after identifying 42 transcripts that are differentially oxidized in bronchial epithelial BEAS-2B cells under air pollution conditions relative to clean air. FDFT1 oxidation affects cholesterol synthesis pathway, leading to phenotypes associated with several lung diseases.
Collapse
|
9
|
Outer membrane vesicles derived from Porphyromonas gingivalis induced cell death with disruption of tight junctions in human lung epithelial cells. Arch Oral Biol 2020; 118:104841. [PMID: 32717445 DOI: 10.1016/j.archoralbio.2020.104841] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Porphyromonas gingivalis (P. gingivalis) is a major bacterium responsible for the progression of periodontitis. P. gingivalis produces small vesicles called outer membrane vesicles (OMVs) containing virulence factors. Increasing evidence suggests a close relationship between periodontitis and respiratory system diseases, such as aspiration pneumonia. However, little is known about whether P. gingivalis OMVs give rise to the impediment of lung epithelial cells. We investigated the effect of the OMVs on cell viability and tight junctions of lung epithelial cells. DESIGN Human lung epithelial A549 cells were treated with P. gingivalis OMVs. Cell viability was evaluated, and cell morphology was examined using scanning electron and phase contrast microscopies. To detect apoptosis induced by P. gingivalis OMVs, activation of caspase-3 and poly ADP-ribose polymerase (PARP) cleavage was examined by using Western blotting. Immunocytochemistry was performed to stain tight junction proteins. RESULTS P. gingivalis OMVs decreased cell viability in A549 cells in a dose- and time-dependent manner. Microscopic analysis revealed that the OMVs induced morphological changes leading to irregular cell membrane structures. The OMVs caused cell shrinkage, membrane blebbing, and cytoplasmic expulsion in a dose-dependent manner. Western blot analysis showed the OMVs induced caspase-3 activation and PARP cleavage. Treatment with the OMVs disrupted the intact distributions of tight junction proteins. CONCLUSIONS These results indicate that P. gingivalis OMVs induced cell death by destroying the barrier system in lung epithelial cells. Our present study raises the possibility that P. gingivalis OMVs is an important factor in the engagement of periodontitis with respiratory system diseases.
Collapse
|
10
|
Ahmadi S, Wang S, Nagpal R, Wang B, Jain S, Razazan A, Mishra SP, Zhu X, Wang Z, Kavanagh K, Yadav H. A human-origin probiotic cocktail ameliorates aging-related leaky gut and inflammation via modulating the microbiota/taurine/tight junction axis. JCI Insight 2020; 5:132055. [PMID: 32302292 DOI: 10.1172/jci.insight.132055] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/08/2020] [Indexed: 12/25/2022] Open
Abstract
Inflammation is a major risk factor of morbidity and mortality in older adults. Although its precise etiology is unknown, low-grade inflammation in older adults is commonly associated with increased intestinal epithelial permeability (leaky gut) and abnormal (dysbiotic) gut microbiota. The increasing older population and lack of treatments to reduce aging-related microbiota dysbiosis, leaky gut, and inflammation culminates in a rise in aging-related comorbidities, constituting a significant public health concern. Here, we demonstrate that a human-origin probiotic cocktail containing 5 Lactobacillus and 5 Enterococcus strains isolated from healthy infant gut prevented high-fat diet-induced (HFD-induced) microbiota dysbiosis, leaky gut, inflammation, metabolic dysfunctions, and physical function decline in older mice. Probiotic-modulated gut microbiota primarily reduced leaky gut by increasing tight junctions, which in turn reduced inflammation. Mechanistically, probiotics modulated microbiota in a way to increase bile salt hydrolase activity, which in turn increased taurine abundance in the gut that stimulated tight junctions and suppressed gut leakiness. Furthermore, in Caenorhabditis elegans, taurine increased life span, reduced adiposity and leaky gut, and enhanced physical function. The results suggest that such probiotic therapies could prevent or treat aging-related leaky gut and inflammation in the elderly.
Collapse
Affiliation(s)
- Shokouh Ahmadi
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Shaohua Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Bo Wang
- Department of Chemistry, North Carolina A&T State University, Greensboro, North Carolina, USA
| | - Shalini Jain
- Department of Internal Medicine-Endocrinology and Metabolism.,Mouse Metabolic Phenotyping Core
| | - Atefeh Razazan
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sidharth P Mishra
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Xuewei Zhu
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| | - Zhan Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Kylie Kavanagh
- Department of Pathology-Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Biomedical Sciences, University of Tasmania, Hobart, Australia
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Microbiology and Immunology, and
| |
Collapse
|
11
|
Fang Q, Hu J, Nie Q, Nie S. Effects of polysaccharides on glycometabolism based on gut microbiota alteration. Trends Food Sci Technol 2019. [DOI: 10.1016/j.tifs.2019.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Abstract
Most new HIV infections, over 80%, occur through sexual transmission. During sexual transmission, the virus must bypass specific female and male reproductive tract anatomical barriers to encounter viable target cells. Understanding the generally efficient ability of these barrier to exclude HIV and the precise mechanisms of HIV translocation beyond these genital barriers is essential for vaccine and novel therapeutic development. In this review, we explore the mucosal, barriers of cervico-vaginal and penile tissues that comprise the female and male reproductive tracts. The unique cellular assemblies f the squamous and columnar epithelium are illustrate highlighting their structure and function. Each anatomical tissue offers a unique barrier to virus entry in healthy individuals. Unfortunately barrier dysfunction can lead to HIV transmission. How these diverse mucosal barriers have the potential to fail is considered, highlighting those anatomical areas that are postulated to offer a weaker barrier and are; therefore, more susceptible to viral ingress. Risk factors, such as sexually transmitted infections, microbiome dysbiosis, and high progestin environments are also associated with increased acquisition of HIV. How these states may affect the integrity of mucosal barriers leading to HIV acquisition are discussed suggesting mechanisms of transmission and revealing potential targets for intervention.
Collapse
Affiliation(s)
- Ann M Carias
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Lurie 9-290, Chicago, IL 60611, USA
| | - Thomas J Hope
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Lurie 9-290, Chicago, IL 60611, USA
| |
Collapse
|
13
|
González-González M, Díaz-Zepeda C, Eyzaguirre-Velásquez J, González-Arancibia C, Bravo JA, Julio-Pieper M. Investigating Gut Permeability in Animal Models of Disease. Front Physiol 2019; 9:1962. [PMID: 30697168 PMCID: PMC6341294 DOI: 10.3389/fphys.2018.01962] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/31/2018] [Indexed: 12/12/2022] Open
Abstract
A growing number of investigations report the association between gut permeability and intestinal or extra-intestinal disorders under the basis that translocation of gut luminal contents could affect tissue function, either directly or indirectly. Still, in many cases it is unknown whether disruption of the gut barrier is a causative agent or a consequence of these conditions. Adequate experimental models are therefore required to further understand the pathophysiology of health disorders associated to gut barrier disruption and to develop and test pharmacological treatments. Here, we review the current animal models that display enhanced intestinal permeability, and discuss (1) their suitability to address mechanistic questions, such as the association between gut barrier alterations and disease and (2) their validity to test potential treatments for pathologies that are characterized by enhanced intestinal permeability.
Collapse
Affiliation(s)
- Marianela González-González
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camilo Díaz-Zepeda
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Johana Eyzaguirre-Velásquez
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camila González-Arancibia
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Javier A Bravo
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| |
Collapse
|
14
|
Herrero R, Prados L, Ferruelo A, Puig F, Pandolfi R, Guillamat-Prats R, Moreno L, Matute-Bello G, Artigas A, Esteban A, Lorente JÁ. Fas activation alters tight junction proteins in acute lung injury. Thorax 2018; 74:69-82. [PMID: 30385692 DOI: 10.1136/thoraxjnl-2018-211535] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 01/11/2023]
Abstract
Background:The acute respiratory distress syndrome (ARDS) is characterized by protein-rich oedema in the alveolar spaces, a feature in which Fas-mediated apoptosis of the alveolar epithelium has been involved. Objective:To determine whether Fas activation increases protein permeability by mechanisms involving disruption of the paracellular tight junction (TJ) proteins in the pulmonary alveoli. Methods: Protein permeability and the expression of TJ proteins were assessed in vivo in wild-type and Fas-deficient lpr mice 16 hours after the intratracheal instillation of recombinant human soluble Fas ligand (rh-sFasL), and at different time points in vitro in human pulmonary alveolar epithelial cells (HPAEpiC) exposed to rh-sFasL Results:Activation of the Fas pathway increased protein permeability in mouse lungs and altered the expression of the TJ proteins occludin and zonula occludens-1 in the alveolar-capillary membrane in vivo and in human alveolar epithelial cell monolayers in vitro. Blockade of caspase-3, but not inhibition of tyrosine kinase dependent pathways, prevented the alterations in TJ protein expression and permeability induced by the Fas/FasL system in human alveolar cell monolayers in vitro. We also observed that both the Fas-induced increase of protein permeability and disruption of TJ proteins occurred before cell death could be detected in the cell monolayers in vitro. Conclusion:Targeting caspase pathways could prevent the disruption of TJs and reduce the formation of lung oedema in the early stages of ARDS.
Collapse
Affiliation(s)
- Raquel Herrero
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Department of Critical Care Medicine, Hospital Universitario de Getafe, Madrid, Spain
| | - Lucia Prados
- Laboratory of Biochemistry, Hospital Universitario de Getafe, Madrid, Spain
| | - Antonio Ferruelo
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain
| | - Ferranda Puig
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Critical Care Center, Corporació Sanitària i Universitària Parc Taul, Institut d' Investigació i Innovació Parc Taulí, Barcelona, Spain
| | - Rachele Pandolfi
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Raquel Guillamat-Prats
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Critical Care Center, Corporació Sanitària i Universitària Parc Taul, Institut d' Investigació i Innovació Parc Taulí, Barcelona, Spain
| | - Laura Moreno
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Gustavo Matute-Bello
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, Department of Medicine, and the Medical Research Service of the VA Puget Sound Healthcare System., University of Washington, Seattle, Washington, USA
| | - Antonio Artigas
- Critical Care Center, Corporació Sanitària i Universitària Parc Taul, Institut d' Investigació i Innovació Parc Taulí, Barcelona, Spain
| | - Andres Esteban
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Universidad Europea, Madrid, Spain
| | - José Ángel Lorente
- CIBER de Enfermedades Respiratorias, Instituto de Investigación Carlos III, Madrid, Spain.,Department of Critical Care Medicine, Hospital Universitario de Getafe, Madrid, Spain.,Universidad Europea, Madrid, Spain
| |
Collapse
|
15
|
Ahmed I, Roy BC, Raach RMT, Owens SM, Xia L, Anant S, Sampath V, Umar S. Enteric infection coupled with chronic Notch pathway inhibition alters colonic mucus composition leading to dysbiosis, barrier disruption and colitis. PLoS One 2018; 13:e0206701. [PMID: 30383855 PMCID: PMC6211731 DOI: 10.1371/journal.pone.0206701] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/12/2018] [Indexed: 12/19/2022] Open
Abstract
Intestinal mucus layer disruption and gut microflora modification in conjunction with tight junction (TJ) changes can increase colonic permeability that allows bacterial dissemination and intestinal and systemic disease. We showed previously that Citrobacter rodentium (CR)-induced colonic crypt hyperplasia and/or colitis is regulated by a functional cross-talk between the Notch and Wnt/β-catenin pathways. In the current study, mucus analysis in the colons of CR-infected (108 CFUs) and Notch blocker Dibenzazepine (DBZ, i.p.; 10μmol/Kg b.w.)-treated mice revealed significant alterations in the composition of trace O-glycans and complex type and hybrid N-glycans, compared to CR-infected mice alone that preceded/accompanied alterations in 16S rDNA microbial community structure and elevated EUB338 staining. While mucin-degrading bacterium, Akkermansia muciniphila (A. muciniphila) along with Enterobacteriaceae belonging to Proteobacteria phyla increased in the feces, antimicrobial peptides Angiogenin-4, Intelectin-1 and Intelectin-2, and ISC marker Dclk1, exhibited dramatic decreases in the colons of CR-infected/DBZ-treated mice. Also evident was a loss of TJ and adherens junction protein immuno-staining within the colonic crypts that negatively impacted paracellular barrier. These changes coincided with the loss of Notch signaling and exacerbation of mucosal injury. In response to a cocktail of antibiotics (Metronidazole/ciprofloxacin) for 10 days, there was increased survival that coincided with: i) decreased levels of Proteobacteria, ii) elevated Dclk1 levels in the crypt and, iii) reduced paracellular permeability. Thus, enteric infections that interfere with Notch activity may promote mucosal dysbiosis that is preceded by changes in mucus composition. Controlled use of antibiotics seems to alleviate gut dysbiosis but may be insufficient to promote colonic crypt regeneration.
Collapse
Affiliation(s)
- Ishfaq Ahmed
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Badal C. Roy
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Rita-Marie T. Raach
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Sarah M. Owens
- Biosciences Division, Argonne National Laboratory, Lemont, Illinois, United States of America
| | - Lijun Xia
- Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center Oklahoma City, Oklahoma, United States of America
| | - Shrikant Anant
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Venkatesh Sampath
- Division of Neonatology, Children’s Mercy Hospital, Kansas City, Missouri, United States of America
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
16
|
Developing a link between toxicants, claudins and neural tube defects. Reprod Toxicol 2018; 81:155-167. [DOI: 10.1016/j.reprotox.2018.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/27/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
|
17
|
Yokouchi M, Kubo A. Maintenance of tight junction barrier integrity in cell turnover and skin diseases. Exp Dermatol 2018; 27:876-883. [DOI: 10.1111/exd.13742] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 02/01/2023]
Affiliation(s)
- Mariko Yokouchi
- Department of Dermatology; Keio University School of Medicine; Tokyo Japan
- Nerima General Hospital; Tokyo Japan
| | - Akiharu Kubo
- Department of Dermatology; Keio University School of Medicine; Tokyo Japan
| |
Collapse
|
18
|
Kuhn KA, Schulz HM, Regner EH, Severs EL, Hendrickson JD, Mehta G, Whitney AK, Ir D, Ohri N, Robertson CE, Frank DN, Campbell EL, Colgan SP. Bacteroidales recruit IL-6-producing intraepithelial lymphocytes in the colon to promote barrier integrity. Mucosal Immunol 2018; 11:357-368. [PMID: 28812548 PMCID: PMC5815964 DOI: 10.1038/mi.2017.55] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/12/2017] [Indexed: 02/04/2023]
Abstract
Interactions between the microbiota and distal gut are important for the maintenance of a healthy intestinal barrier; dysbiosis of intestinal microbial communities has emerged as a likely contributor to diseases that arise at the level of the mucosa. Intraepithelial lymphocytes (IELs) are positioned within the epithelial barrier, and in the small intestine they function to maintain epithelial homeostasis. We hypothesized that colon IELs promote epithelial barrier function through the expression of cytokines in response to interactions with commensal bacteria. Profiling of bacterial 16S ribosomal RNA revealed that candidate bacteria in the order Bacteroidales are sufficient to promote IEL presence in the colon that in turn produce interleukin-6 (IL-6) in a MyD88 (myeloid differentiation primary response 88)-dependent manner. IEL-derived IL-6 is functionally important in the maintenance of the epithelial barrier as IL-6-/- mice were noted to have increased paracellular permeability, decreased claudin-1 expression, and a thinner mucus gel layer, all of which were reversed by transfer of IL-6+/+ IELs, leading to protection of mice in response to Citrobacter rodentium infection. Therefore, we conclude that microbiota provide a homeostatic role for epithelial barrier function through regulation of IEL-derived IL-6.
Collapse
Affiliation(s)
- Kristine A. Kuhn
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Hanna M. Schulz
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Emilie H. Regner
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
| | - Erin L. Severs
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Jason D. Hendrickson
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Gaurav Mehta
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Alyssa K. Whitney
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Diana Ir
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Neha Ohri
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Charles E. Robertson
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Daniel N. Frank
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Eric L. Campbell
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Sean P. Colgan
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| |
Collapse
|
19
|
|
20
|
Kim Y, Kessler SP, Obery DR, Homer CR, McDonald C, de la Motte CA. Hyaluronan 35kDa treatment protects mice from Citrobacter rodentium infection and induces epithelial tight junction protein ZO-1 in vivo. Matrix Biol 2017; 62:28-39. [PMID: 27845198 PMCID: PMC5427001 DOI: 10.1016/j.matbio.2016.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 12/20/2022]
Abstract
Maintaining a healthy intestinal barrier, the primary physical barrier between intestinal microbiota and the underlying lamina propria, is critical for optimal health. Epithelial integrity is essential for the prevention of the entrance of luminal contents, such as bacteria and their products, through the large intestinal barrier. In this study, we investigated the protective functions of biosynthetic, specific sized, hyaluronan around 35kDa (HA35) on intestinal epithelium in healthy mice, as well as mice infected Citrobacter rodentium, an established model that mimics infection with a serious human pathogen, enteropathogenic E. coli (EPEC). Our results reveal that treatment with HA35 protects mice from Citrobacter infection and enhances the epithelial barrier function. In particular, we have found that HA35 induces the expression of tight junction protein zonula occludens (ZO)-1 in both healthy and Citrobacter infected mice, as demonstrated by immunoflurorescence and Western blot analyses. Furthermore, we determined that HA35 treatment enhances ZO-1 expression and reduces intestinal permeability at the early stages of dextran sulfate sodium (DSS)-induced colitis in mice. Together, our data demonstrate that the expression and functionality of tight junctions, are increased by HA35 treatment, suggesting a novel mechanism for the protection from Citrobacter infection.
Collapse
Affiliation(s)
- Yeojung Kim
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sean P Kessler
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Dana R Obery
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Craig R Homer
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Christine McDonald
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Carol A de la Motte
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
21
|
Khadilkar RJ, Vogl W, Goodwin K, Tanentzapf G. Modulation of occluding junctions alters the hematopoietic niche to trigger immune activation. eLife 2017; 6:28081. [PMID: 28841136 PMCID: PMC5597334 DOI: 10.7554/elife.28081] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/24/2017] [Indexed: 12/04/2022] Open
Abstract
Stem cells are regulated by signals from their microenvironment, or niche. During Drosophila hematopoiesis, a niche regulates prohemocytes to control hemocyte production. Immune challenges activate cell-signalling to initiate the cellular and innate immune response. Specifically, certain immune challenges stimulate the niche to produce signals that induce prohemocyte differentiation. However, the mechanisms that promote prohemocyte differentiation subsequent to immune challenges are poorly understood. Here we show that bacterial infection induces the cellular immune response by modulating occluding-junctions at the hematopoietic niche. Occluding-junctions form a permeability barrier that regulates the accessibility of prohemocytes to niche derived signals. The immune response triggered by infection causes barrier breakdown, altering the prohemocyte microenvironment to induce immune cell production. Moreover, genetically induced barrier ablation provides protection against infection by activating the immune response. Our results reveal a novel role for occluding-junctions in regulating niche-hematopoietic progenitor signalling and link this mechanism to immune cell production following infection.
Collapse
Affiliation(s)
- Rohan J Khadilkar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Wayne Vogl
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Katharine Goodwin
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
22
|
The basolateral vesicle sorting machinery and basolateral proteins are recruited to the site of enteropathogenic E. coli microcolony growth at the apical membrane. PLoS One 2017. [PMID: 28636623 PMCID: PMC5479554 DOI: 10.1371/journal.pone.0179122] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Foodborne Enteropathogenic Escherichia coli (EPEC) infections of the small intestine cause diarrhea especially in children and are a major cause of childhood death in developing countries. EPEC infects the apical membrane of the epithelium of the small intestine by attaching, effacing the microvilli under the bacteria and then forming microcolonies on the cell surface. We first asked the question where on epithelial cells EPEC attaches and grows. Using models of polarized epithelial monolayers, we evaluated the sites of initial EPEC attachment to the apical membrane and found that EPEC preferentially attached over the cell-cell junctions and formed microcolonies preferentially where three cells come together at tricellular tight junctions. The ability of EPEC to adhere increased when host cell polarity was compromised yielding EPEC access to basolateral proteins. EPEC pedestals contain basolateral cytoskeletal proteins. Thus, we asked if attached EPEC causes reorganization the protein composition of the host cell plasma membrane at sites of microcolony formation. We found that EPEC microcolony growth at the apical membrane resulted in a local accumulation of basolateral plasma membrane proteins surrounding the microcolony. Basolateral marker protein aquaporin-3 localized to forming EPEC microcolonies. Components of the basolateral vesicle targeting machinery were re-routed. The Exocyst (Exo70) was recruited to individual EPEC as was the basolateral vesicle SNARE VAMP-3. Moreover, several Rab variants were also recruited to the infection site, and their dominant-negative equivalents were not. To quantitatively study the recruitment of basolateral proteins, we created a pulse of the temperature sensitive basolateral VSVG, VSVG3-SP-GFP, from the trans-Golgi Network. We found that after release from the TGN, significantly more VSVG3-SP-GFP accumulated at the site of microcolony growth than on equivalent membrane regions of uninfected cells. This suggests that trafficking of vesicles destined for the basolateral membrane are redirected to the apical site of microcolony growth. Thus, in addition to disrupting host cell fence function, local host cell plasma membrane protein composition is changed by altered protein trafficking and recruitment of basolateral proteins to the apical microcolony. This may aid EPEC attachment and subsequent microcolony growth.
Collapse
|
23
|
Ahmad R, Sorrell MF, Batra SK, Dhawan P, Singh AB. Gut permeability and mucosal inflammation: bad, good or context dependent. Mucosal Immunol 2017; 10:307-317. [PMID: 28120842 PMCID: PMC6171348 DOI: 10.1038/mi.2016.128] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/29/2016] [Indexed: 02/04/2023]
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease. A breach in the mucosal barrier, otherwise known as "leaky gut," is alleged to promote mucosal inflammation by intensifying immune activation. However, interaction between the luminal antigen and mucosal immune system is necessary to maintain mucosal homeostasis. Furthermore, manipulations leading to deregulated gut permeability have resulted in susceptibility in mice to colitis as well as to creating adaptive immunity. These findings implicate a complex but dynamic association between mucosal permeability and immune homeostasis; however, they also emphasize that compromised gut permeability alone may not be sufficient to induce colitis. Emerging evidence further supports the role(s) of proteins associated with the mucosal barrier in epithelial injury and repair: manipulations of associated proteins also modified epithelial differentiation, proliferation, and apoptosis. Taken together, the role of gut permeability and proteins associated in regulating mucosal inflammatory diseases appears to be more complex than previously thought. Herein, we review outcomes from recent mouse models where gut permeability was altered by direct and indirect effects of manipulating mucosal barrier-associated proteins, to highlight the significance of mucosal permeability and the non-barrier-related roles of these proteins in regulating chronic mucosal inflammatory conditions.
Collapse
Affiliation(s)
- R Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, Nebraska, USA
| | - MF Sorrell
- Department of Internal Medicine, Omaha, Nebraska, USA
| | - SK Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, Nebraska, USA.,Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska USA and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska USA
| | - P Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, Nebraska, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska USA and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska USA.,VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska USA
| | - AB Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, Nebraska, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska USA and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska USA.,VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska USA
| |
Collapse
|
24
|
Biffi E. Microbiome and Cardiac Health. INTEGRATIVE CARDIOLOGY 2017:67-97. [DOI: 10.1007/978-3-319-40010-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
25
|
Targeting and alteration of tight junctions by bacteria and their virulence factors such as Clostridium perfringens enterotoxin. Pflugers Arch 2016; 469:77-90. [DOI: 10.1007/s00424-016-1902-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/01/2016] [Accepted: 11/06/2016] [Indexed: 01/01/2023]
|
26
|
Evaluation of therapeutic properties of fermented vegetables extract (OM-X®) in the model of colitis induced by Citrobacter rodentium in mice. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
27
|
Abstract
Citrobacter rodentium is a mucosal pathogen of mice that shares several pathogenic mechanisms with enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC), which are two clinically important human gastrointestinal pathogens. Thus, C. rodentium has long been used as a model to understand the molecular basis of EPEC and EHEC infection in vivo. In this Review, we discuss recent studies in which C. rodentium has been used to study mucosal immunology, including the deregulation of intestinal inflammatory responses during bacteria-induced colitis and the role of the intestinal microbiota in mediating resistance to colonization by enteric pathogens. These insights should help to elucidate the roles of mucosal inflammatory responses and the microbiota in the virulence of enteric pathogens.
Collapse
|
28
|
Mallick EM, Garber JJ, Vanguri VK, Balasubramanian S, Blood T, Clark S, Vingadassalom D, Louissaint C, McCormick B, Snapper SB, Leong JM. The ability of an attaching and effacing pathogen to trigger localized actin assembly contributes to virulence by promoting mucosal attachment. Cell Microbiol 2014; 16:1405-24. [PMID: 24780054 DOI: 10.1111/cmi.12302] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/09/2014] [Accepted: 04/13/2014] [Indexed: 12/30/2022]
Abstract
Enterohaemorrhagic Escherichia coli (EHEC) colonizes the intestine and causes bloody diarrhoea and kidney failure by producing Shiga toxin. Upon binding intestinal cells, EHEC triggers a change in host cell shape, generating actin 'pedestals' beneath bound bacteria. To investigate the importance of pedestal formation to disease, we infected genetically engineered mice incapable of supporting pedestal formation by an EHEC-like mouse pathogen, or wild type mice with a mutant of that pathogen incapable of generating pedestals. We found that pedestal formation promotes attachment of bacteria to the intestinal mucosa and vastly increases the severity of Shiga toxin-mediated disease.
Collapse
Affiliation(s)
- Emily M Mallick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Beatty JK, Bhargava A, Buret AG. Post-infectious irritable bowel syndrome: Mechanistic insights into chronic disturbances following enteric infection. World J Gastroenterol 2014; 20:3976-3985. [PMID: 24744587 PMCID: PMC3983453 DOI: 10.3748/wjg.v20.i14.3976] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/09/2014] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a commonly encountered chronic functional gastrointestinal (GI) disorder. Approximately 10% of IBS patients can trace the onset of their symptoms to a previous a bout of infectious dysentery. The appearance of new IBS symptoms following an infectious event is defined as post-infectious-IBS. Indeed, with the World Health Organization estimating between 2 and 4 billion cases annually, infectious diarrheal disease represents an incredible international healthcare burden. Additionally, compounding evidence suggests many commonly encountered enteropathogens as unique triggers behind IBS symptom generation and underlying pathophysiological features. A growing body of work provides evidence supporting a role for pathogen-mediated modifications in the resident intestinal microbiota, epithelial barrier integrity, effector cell functions, and innate and adaptive immune features, all proposed physiological manifestations that can underlie GI abnormalities in IBS. Enteric pathogens must employ a vast array of machinery to evade host protective immune mechanisms, and illicit successful infections. Consequently, the impact of infectious events on host physiology can be multidimensional in terms of anatomical location, functional scope, and duration. This review offers a unique discussion of the mechanisms employed by many commonly encountered enteric pathogens that cause acute disease, but may also lead to the establishment of chronic GI dysfunction compatible with IBS.
Collapse
|
30
|
Anderson RC, Young W, Clerens S, Cookson AL, McCann MJ, Armstrong KM, Roy NC. Human oral isolate Lactobacillus fermentum AGR1487 reduces intestinal barrier integrity by increasing the turnover of microtubules in Caco-2 cells. PLoS One 2013; 8:e78774. [PMID: 24244356 PMCID: PMC3828418 DOI: 10.1371/journal.pone.0078774] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/16/2013] [Indexed: 02/06/2023] Open
Abstract
Lactobacillus fermentum is found in fermented foods and thought to be harmless. In vivo and clinical studies indicate that some L. fermentum strains have beneficial properties, particularly for gastrointestinal health. However, L. fermentum AGR1487 decreases trans-epithelial electrical resistance (TEER), a measure of intestinal barrier integrity. The hypothesis was that L. fermentum AGR1487 decreases the expression of intestinal cell tight junction genes and proteins, thereby reducing barrier integrity. Transcriptomic and proteomic analyses of Caco-2 cells (model of human intestinal epithelial cells) treated with L. fermentum AGR1487 were used to obtain a global view of the effect of the bacterium on intestinal epithelial cells. Specific functional characteristics by which L. fermentum AGR1487 reduces intestinal barrier integrity were examined using confocal microscopy, cell cycle progression and adherence bioassays. The effects of TEER-enhancing L. fermentum AGR1485 were investigated for comparison. L. fermentum AGR1487 did not alter the expression of Caco-2 cell tight junction genes (compared to L. fermentum AGR1485) and tight junction proteins were not able to be detected. However, L. fermentum AGR1487 increased the expression levels of seven tubulin genes and the abundance of three microtubule-associated proteins, which have been linked to tight junction disassembly. Additionally, Caco-2 cells treated with L. fermentum AGR1487 did not have defined and uniform borders of zona occludens 2 around each cell, unlike control or AGR1485 treated cells. L. fermentum AGR1487 cells were required for the negative effect on barrier integrity (bacterial supernatant did not cause a decrease in TEER), suggesting that a physical interaction may be necessary. Increased adherence of L. fermentum AGR1487 to Caco-2 cells (compared to L. fermentum AGR1485) was likely to facilitate this cell-to-cell interaction. These findings illustrate that bacterial strains of the same species can cause contrasting host responses and suggest that food-safe status should be given to individual strains not species.
Collapse
Affiliation(s)
- Rachel C. Anderson
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- * E-mail:
| | - Wayne Young
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Stefan Clerens
- Proteins & Biomaterials Team, Food & Bio-based Products Group, AgResearch Lincoln, Lincoln, New Zealand
| | - Adrian L. Cookson
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Rumen Microbiology Team, Animal Health & Nutrition Group, AgResearch Grasslands, Palmerston North, New Zealand
| | - Mark J. McCann
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Gravida: National Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| | - Kelly M. Armstrong
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
| | - Nicole C. Roy
- Food Nutrition & Health Team, Food & Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
- Rumen Microbiology Team, Animal Health & Nutrition Group, AgResearch Grasslands, Palmerston North, New Zealand
- Gravida: National Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Che P, Tang H, Li Q. The interaction between claudin-1 and dengue viral prM/M protein for its entry. Virology 2013; 446:303-13. [PMID: 24074594 DOI: 10.1016/j.virol.2013.08.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/18/2013] [Accepted: 08/09/2013] [Indexed: 02/04/2023]
Abstract
Dengue disease is becoming a huge public health concern around the world as more than one-third of the world's population living in areas at risk of infection. In an effort to assess host factors interacting with dengue virus, we identified claudin-1, a major tight junction component, as an essential cell surface protein for dengue virus entry. When claudin-1 was knocked down in Huh 7.5 cells via shRNA, the amount of dengue virus entering host cells was reduced. Consequently, the progeny virus productions were decreased and dengue virus-induced CPE was prevented. Furthermore, restoring the expression of claudin-1 in the knockdown cells facilitated dengue virus entry. The interaction between claudin-1 and dengue viral prM protein was further demonstrated using the pull-down assay. Deletion of the extracellular loop 1 (ECL1) of claudin-1 abolished such interaction, so did point mutations C54A, C64A and I32M on ECL1. These results suggest that the interaction between viral protein prM and host protein claudin-1 was essential for dengue entry. Since host and viral factors involved in virus entry are promising therapeutic targets, determining the essential role of claudin-1 could lead to the discovery of entry inhibitors with attractive therapeutic potential against dengue disease.
Collapse
Affiliation(s)
- Pulin Che
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; University of Alabama at Birmingham, Department of Medicine, Division of Infectious Diseases, BBRB 562, 845 19th Street South, Birmingham, AL 35294, United States
| | | | | |
Collapse
|
32
|
Tsai YH, Disson O, Bierne H, Lecuit M. Murinization of internalin extends its receptor repertoire, altering Listeria monocytogenes cell tropism and host responses. PLoS Pathog 2013; 9:e1003381. [PMID: 23737746 PMCID: PMC3667765 DOI: 10.1371/journal.ppat.1003381] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 04/09/2013] [Indexed: 01/17/2023] Open
Abstract
Listeria monocytogenes (Lm) is an invasive foodborne pathogen that leads to severe central nervous system and maternal-fetal infections. Lm ability to actively cross the intestinal barrier is one of its key pathogenic properties. Lm crosses the intestinal epithelium upon the interaction of its surface protein internalin (InlA) with its host receptor E-cadherin (Ecad). InlA-Ecad interaction is species-specific, does not occur in wild-type mice, but does in transgenic mice expressing human Ecad and knock-in mice expressing humanized mouse Ecad. To study listeriosis in wild-type mice, InlA has been “murinized” to interact with mouse Ecad. Here, we demonstrate that, unexpectedly, murinized InlA (InlAm) mediates not only Ecad-dependent internalization, but also N-cadherin-dependent internalization. Consequently, InlAm-expressing Lm targets not only goblet cells expressing luminally-accessible Ecad, as does Lm in humanized mice, but also targets villous M cells, which express luminally-accessible N-cadherin. This aberrant Lm portal of entry results in enhanced innate immune responses and intestinal barrier damage, both of which are not observed in wild-type Lm-infected humanized mice. Murinization of InlA therefore not only extends the host range of Lm, but also broadens its receptor repertoire, providing Lm with artifactual pathogenic properties. These results challenge the relevance of using InlAm-expressing Lm to study human listeriosis and in vivo host responses to this human pathogen. Co-evolution of microbes with their hosts can select stringently specific host-microbe interactions at the cell, tissue and species levels. Listeria monocytogenes (Lm) is a foodborne pathogen that causes a deadly systemic infection in humans. Lm crosses the intestinal epithelium upon the interaction of its surface protein InlA with E-cadherin (Ecad). InlA-Ecad interaction is species-specific, does not occur in wild-type mice, but does in transgenic mice expressing human Ecad and knock-in mice expressing humanized mouse Ecad. To study listeriosis in wild-type mice, InlA has been “murinized” to interact with mouse Ecad. Here, we demonstrate that in addition to interacting with mouse Ecad, InlAm also uses N-cadherin as a receptor, whereas InlA does not. This artifactual InlAm-N-cadherin interaction promotes bacterial translocation across villous M cells, a cell type which is not targeted by InlA-expressing bacteria. This leads to intestinal inflammation and intestinal barrier damage, both of which are not seen in humans and humanized mouse models permissive to InlA-Ecad interaction. These results challenge the relevance of using InlAm-expressing Lm as a model to study human listeriosis and host responses to this pathogen. They also illustrate that caution must be exercised before using “murinized” pathogens to study human infectious diseases.
Collapse
Affiliation(s)
- Yu-Huan Tsai
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Olivier Disson
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
| | - Hélène Bierne
- Institut Pasteur, Unité des Interactions Bactéries Cellules, Paris, France
- Inserm, U604, Paris, France
- INRA, USC2020, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France
- Inserm U1117, Paris, France
- Institut Pasteur, French National Reference Center and World Health Organization Collaborating Center on Listeria, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
- Necker-Enfants Malades University Hospital, APHP, Division of Infectious Diseases and Tropical Medicine, Paris, France
- * E-mail:
| |
Collapse
|
33
|
Li C, Wang R, Su B, Luo Y, Terhune J, Beck B, Peatman E. Evasion of mucosal defenses during Aeromonas hydrophila infection of channel catfish (Ictalurus punctatus) skin. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 39:447-455. [PMID: 23219904 DOI: 10.1016/j.dci.2012.11.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/20/2012] [Accepted: 11/20/2012] [Indexed: 06/01/2023]
Abstract
The mucosal surfaces of fish serve as the first line of defense against the myriad of aquatic pathogens present in the aquatic environment. The immune repertoire functioning at these interfaces is still poorly understood. The skin, in particular, must process signals from several fronts, sensing and integrating environmental, nutritional, social, and health cues. Pathogen invasion can disrupt this delicate homeostasis with profound impacts on signaling throughout the organism. Here, we investigated the transcriptional effects of virulent Aeromonas hydrophila infection in channel catfish skin, Ictalurus punctatus. We utilized a new 8 × 60 K Agilent microarray for catfish to examine gene expression profiles at critical early timepoints following challenge--2 h, 8 h, and 12 h. Expression of a total of 2,168 unique genes was significantly perturbed during at least one timepoint. We observed dysregulation of genes involved in antioxidant, cytoskeletal, immune, junctional, and nervous system pathways. In particular, A. hydrophila infection rapidly altered a number of potentially critical lectins, chemokines, interleukins, and other mucosal factors in a manner predicted to enhance its ability to adhere to and invade the catfish host.
Collapse
Affiliation(s)
- Chao Li
- Department of Fisheries and Allied Aquacultures, Auburn University, Auburn, AL 36849, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Buret AG, Bhargava A. Modulatory mechanisms of enterocyte apoptosis by viral, bacterial and parasitic pathogens. Crit Rev Microbiol 2013; 40:1-17. [PMID: 23297858 DOI: 10.3109/1040841x.2012.746952] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Abstract
The intestinal barrier serves 2 critical functions for the survival of the individual: first, it allows nutrient absorption and second, it defends the body from dangerous macromolecule penetration. It is a complex multilayer system, consisting of an external "anatomic" barrier and an inner "functional" immunological barrier. The interaction of these 2 barriers enables equilibrated permeability to be maintained. Many factors can alter this balance: gut microflora modifications, mucus layer alterations, and epithelial damage can increase intestinal permeability, allowing the translocation of luminal content to the inner layer of intestinal wall. Several techniques are now available that enable us to study gut permeability: "in vitro" models (Caco-2 and HT29-MTX cells) and "in vivo" not invasive tests (sugar tests and radioisotope scanning tests) are used to estimate permeability and to suggest molecular pathophysiological mechanisms of intestinal permeability in health and diseases. Many medicinal products used in the treatment of gastrointestinal diseases have also found to play an active role in modulate intestinal permeability: corticosteroids, 5-aminosalicylic acid, anti-tumor necrosis factor, probiotics, and mucosal protectors, like gelatin tannate. This review will particularly address the role of the gut barrier in maintaining intestinal permeability (microbiota, mucus, and epithelial cells), the techniques used for estimating intestinal permeability and the therapeutic approaches able to modify it.
Collapse
|
36
|
Zhang Q, Li Q, Wang C, Li N, Li J. Redistribution of tight junction proteins during EPEC infection in vivo. Inflammation 2012; 35:23-32. [PMID: 21170673 DOI: 10.1007/s10753-010-9285-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC) is a leading cause of diarrhea among infants. Tight junction plays a vital role in intestinal paracellular permeability by forming physical intercellular barriers in epithelial cells. However, the impact of this enteric pathogen on tight junctions in vivo has not been fully investigated. In the present study, the alterations in tight junctions following EPEC infection in vivo were investigated. Western blot analysis revealed that the tight junction proteins, occludin and claudin-1, were displaced from tight junction membrane microdomains to Triton X-100 soluble fractions after EPEC infection. Changes in intestinal paracellular permeability were determined using the molecular tracer biotin, which was observed to penetrate the epithelia and extended into the lamina propria, indicating disruption in tight junction barrier function. Our results suggested that redistribution of tight junction proteins plays an important role in the disruption of epithelial barrier function induced by EPEC infection, which may provide new insight into the pathogenesis of diarrhea caused by EPEC.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, People's Republic of China
| | | | | | | | | |
Collapse
|
37
|
Ritchie JM, Rui H, Zhou X, Iida T, Kodoma T, Ito S, Davis BM, Bronson RT, Waldor MK. Inflammation and disintegration of intestinal villi in an experimental model for Vibrio parahaemolyticus-induced diarrhea. PLoS Pathog 2012; 8:e1002593. [PMID: 22438811 PMCID: PMC3305451 DOI: 10.1371/journal.ppat.1002593] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 02/01/2012] [Indexed: 12/17/2022] Open
Abstract
Vibrio parahaemolyticus is a leading cause of seafood-borne gastroenteritis in many parts of the world, but there is limited knowledge of the pathogenesis of V. parahaemolyticus-induced diarrhea. The absence of an oral infection-based small animal model to study V. parahaemolyticus intestinal colonization and disease has constrained analyses of the course of infection and the factors that mediate it. Here, we demonstrate that infant rabbits oro-gastrically inoculated with V. parahaemolyticus develop severe diarrhea and enteritis, the main clinical and pathologic manifestations of disease in infected individuals. The pathogen principally colonizes the distal small intestine, and this colonization is dependent upon type III secretion system 2. The distal small intestine is also the major site of V. parahaemolyticus-induced tissue damage, reduced epithelial barrier function, and inflammation, suggesting that disease in this region of the gastrointestinal tract accounts for most of the diarrhea that accompanies V. parahaemolyticus infection. Infection appears to proceed through a characteristic sequence of steps that includes remarkable elongation of microvilli and the formation of V. parahaemolyticus-filled cavities within the epithelial surface, and culminates in villus disruption. Both depletion of epithelial cell cytoplasm and epithelial cell extrusion contribute to formation of the cavities in the epithelial surface. V. parahaemolyticus also induces proliferation of epithelial cells and recruitment of inflammatory cells, both of which occur before wide-spread damage to the epithelium is evident. Collectively, our findings suggest that V. parahaemolyticus damages the host intestine and elicits disease via previously undescribed processes and mechanisms. The marine bacterium Vibrio parahaemolyticus is a leading cause worldwide of gastroenteritis linked to the consumption of contaminated seafood. Despite the prevalence of V. parahaemolyticus-induced gastroenteritis, there is limited understanding of how this pathogen causes disease in the intestine. In part, the paucity of knowledge results from the absence of an oral infection-based animal model of the human disease. We developed a simple oral infection-based infant rabbit model of V. parahaemolyticus-induced intestinal pathology and diarrhea. This experimental model enabled us to define several previously unknown but key features of the pathology elicited by this organism. We found that V. parahaemolyticus chiefly colonizes the distal small intestine and that the organism's second type III secretion system is essential for colonization. The epithelial surface of the distal small intestine is also the major site of V. parahaemolyticus-induced damage, which arises via a characteristic sequence of events culminating in the formation of V. parahaemolyticus-filled cavities in the epithelial surface. This experimental model will transform future studies aimed at deciphering the bacterial and host factors/processes that contribute to disease, as well as enable testing of new therapeutics to prevent and/or combat infection.
Collapse
Affiliation(s)
- Jennifer M. Ritchie
- Brigham and Women's Hospital/Harvard Medical School and HHMI, Boston, Massachusetts, United States of America
- * E-mail: (MKW); (JMR)
| | - Haopeng Rui
- Brigham and Women's Hospital/Harvard Medical School and HHMI, Boston, Massachusetts, United States of America
| | - Xiaohui Zhou
- Brigham and Women's Hospital/Harvard Medical School and HHMI, Boston, Massachusetts, United States of America
| | - Tetsuya Iida
- Department of Bacterial Infections, International Research Center for Infectious Diseases, Osaka University, Suita, Osaka, Japan
| | - Toshio Kodoma
- Laboratory of Genomic Research on Pathogenic Bacteria, International Research Center for Infectious Diseases, Osaka University, Suita, Osaka, Japan
| | - Susuma Ito
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brigid M. Davis
- Brigham and Women's Hospital/Harvard Medical School and HHMI, Boston, Massachusetts, United States of America
| | - Roderick T. Bronson
- Department of Microbiology & Immunology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Matthew K. Waldor
- Brigham and Women's Hospital/Harvard Medical School and HHMI, Boston, Massachusetts, United States of America
- * E-mail: (MKW); (JMR)
| |
Collapse
|
38
|
Khan MW, Kale AA, Bere P, Vajjala S, Gounaris E, Pakanati KC. Microbes, intestinal inflammation and probiotics. Expert Rev Gastroenterol Hepatol 2012; 6:81-94. [PMID: 22149584 DOI: 10.1586/egh.11.94] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Inflammatory bowel disease (IBD) is known for causing disturbed homeostatic balance among the intestinal immune compartment, epithelium and microbiota. Owing to the emergence of IBD as a major cause of morbidity and mortality, great efforts have been put into understanding the sequence of intestinal inflammatory events. Intestinal macrophages and dendritic cells act in a synergistic fashion with intestinal epithelial cells and microbiota to initiate the triad that governs the intestinal immune responses (whether inflammatory or regulatory). In this review, we will discuss the interplay of intestinal epithelial cells, bacteria and the innate immune component. Moreover, whether or not genetic intervention of probiotic bacteria is a valid approach for attenuating/mitigating exaggerated inflammation and IBD will also be discussed.
Collapse
Affiliation(s)
- Mohammad W Khan
- The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Bonazzi M, Cossart P. Impenetrable barriers or entry portals? The role of cell-cell adhesion during infection. ACTA ACUST UNITED AC 2012; 195:349-58. [PMID: 22042617 PMCID: PMC3206337 DOI: 10.1083/jcb.201106011] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell-cell adhesion plays a fundamental role in cell polarity and organogenesis. It also contributes to the formation and establishment of physical barriers against microbial infections. However, a large number of pathogens, from viruses to bacteria and parasites, have developed countless strategies to specifically target cell adhesion molecules in order to adhere to and invade epithelial cells, disrupt epithelial integrity, and access deeper tissues for dissemination. The study of all these processes has contributed to the characterization of molecular machineries at the junctions of eukaryotic cells that have been better understood by using pathogens as probes.
Collapse
Affiliation(s)
- Matteo Bonazzi
- Centre Nationale de la Recherche Scientifique, UMR 5236, CPBS, CNRS, 34293 Montpellier, France.
| | | |
Collapse
|
40
|
Conterno L, Fava F, Viola R, Tuohy KM. Obesity and the gut microbiota: does up-regulating colonic fermentation protect against obesity and metabolic disease? GENES & NUTRITION 2011; 6:241-60. [PMID: 21559992 PMCID: PMC3145060 DOI: 10.1007/s12263-011-0230-1] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 04/20/2011] [Indexed: 12/18/2022]
Abstract
Obesity is now considered a major public health concern globally as it predisposes to a number of chronic human diseases. Most developed countries have experienced a dramatic and significant rise in obesity since the 1980s, with obesity apparently accompanying, hand in hand, the adoption of "Western"-style diets and low-energy expenditure lifestyles around the world. Recent studies report an aberrant gut microbiota in obese subjects and that gut microbial metabolic activities, especially carbohydrate fermentation and bile acid metabolism, can impact on a number of mammalian physiological functions linked to obesity. The aim of this review is to present the evidence for a characteristic "obese-type" gut microbiota and to discuss studies linking microbial metabolic activities with mammalian regulation of lipid and glucose metabolism, thermogenesis, satiety, and chronic systemic inflammation. We focus in particular on short-chain fatty acids (SCFA) produced upon fiber fermentation in the colon. Although SCFA are reported to be elevated in the feces of obese individuals, they are also, in contradiction, identified as key metabolic regulators of the physiological checks and controls mammals rely upon to regulate energy metabolism. Most studies suggest that the gut microbiota differs in composition between lean and obese individuals and that diet, especially the high-fat low-fiber Western-style diet, dramatically impacts on the gut microbiota. There is currently no consensus as to whether the gut microbiota plays a causative role in obesity or is modulated in response to the obese state itself or the diet in obesity. Further studies, especially on the regulatory role of SCFA in human energy homeostasis, are needed to clarify the physiological consequences of an "obese-style" microbiota and any putative dietary modulation of associated disease risk.
Collapse
Affiliation(s)
- Lorenza Conterno
- Nutrition and Nutrigenomics Group, Research and Innovation Centre, FEM-IASMA, 38010, S. Michele a.A, Trento, Italy,
| | | | | | | |
Collapse
|
41
|
Abstract
This review discusses the barriers an enteric pathogen encounters when establishing an infection in the intestinal tract. There are potential barriers in the lumen that increase competition for nutrients and space. The role of mucus layer, and the antimicrobial peptides and secretory IgA sequestered within it, are also significant barriers. After overcoming these defences, the pathogen encounters the epithelial layer. This layer can be broken down into various protective components including enterocytes, Paneth cells, goblet cells, M cells and pathogen recognition receptors. Collectively, these intestinal defences constitute significant barriers that pathogens must overcome to successfully colonize this important mucosal surface.
Collapse
Affiliation(s)
- Navkiran Gill
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | | | | |
Collapse
|
42
|
Anderson RC, Cookson AL, McNabb WC, Park Z, McCann MJ, Kelly WJ, Roy NC. Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol 2010; 10:316. [PMID: 21143932 PMCID: PMC3004893 DOI: 10.1186/1471-2180-10-316] [Citation(s) in RCA: 325] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 12/09/2010] [Indexed: 02/07/2023] Open
Abstract
Background Intestinal barrier function is important for preserving health, as a compromised barrier allows antigen entry and can induce inflammatory diseases. Probiotic bacteria can play a role in enhancing intestinal barrier function; however, the mechanisms are not fully understood. Existing studies have focused on the ability of probiotics to prevent alterations to tight junctions in disease models, and have been restricted to a few tight junction bridging proteins. No studies have previously investigated the effect of probiotic bacteria on healthy intestinal epithelial cell genes involved in the whole tight junction signalling pathway, including those encoding for bridging, plaque and dual location tight junction proteins. Alteration of tight junction signalling in healthy humans is a potential mechanism that could lead to the strengthening of the intestinal barrier, resulting in limiting the ability of antigens to enter the body and potentially triggering undesirable immune responses. Results The effect of Lactobacillus plantarum MB452 on tight junction integrity was determined by measuring trans-epithelial electrical resistance (TEER) across Caco-2 cell layers. L. plantarum MB452 caused a dose-dependent TEER increase across Caco-2 cell monolayers compared to control medium. Gene expression was compared in Caco-2 cells untreated or treated with L. plantarum MB452 for 10 hours. Caco-2 cell RNA was hybridised to human oligonucleotide arrays. Data was analysed using linear models and differently expressed genes were examined using pathway analysis tools. Nineteen tight junction-related genes had altered expression levels in response to L. plantarum MB452 (modified-P < 0.05, fold-change > 1.2), including those encoding occludin and its associated plaque proteins that anchor it to the cytoskeleton. L. plantarum MB452 also caused changes in tubulin and proteasome gene expression levels which may be linked to intestinal barrier function. Caco-2 tight junctions were visualised by fluorescent microscopy of immuno-stained occludin, zona occludens (ZO)-1, ZO-2 and cingulin. Caco-2 cells treated with L. plantarum MB452 had higher intensity fluorescence of each of the four tight junction proteins compared to untreated controls. Conclusions This research indicates that enhancing the expression of genes involved in tight junction signalling is a possible mechanism by which L. plantarum MB452 improves intestinal barrier function.
Collapse
Affiliation(s)
- Rachel C Anderson
- AgriFoods & Health Section, Food & Textiles Group, AgResearch Grasslands, Private Bag 11008, Palmerston North 4442, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
43
|
Kim M, Ashida H, Ogawa M, Yoshikawa Y, Mimuro H, Sasakawa C. Bacterial interactions with the host epithelium. Cell Host Microbe 2010; 8:20-35. [PMID: 20638639 DOI: 10.1016/j.chom.2010.06.006] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 04/03/2010] [Accepted: 04/27/2010] [Indexed: 12/14/2022]
Abstract
The gastrointestinal epithelium deploys multiple innate defense mechanisms to fight microbial intruders, including epithelial integrity, rapid epithelial cell turnover, quick expulsion of infected cells, autophagy, and innate immune responses. Nevertheless, many bacterial pathogens are equipped with highly evolved infectious stratagems that circumvent these defense systems and use the epithelium as a replicative foothold. During replication on and within the gastrointestinal epithelium, gastrointestinal bacterial pathogens secrete various components, toxins, and effectors that can subvert, usurp, and exploit host cellular functions to benefit bacterial survival. In addition, bacterial pathogens use a variety of mechanisms that balance breaching the epithelial barrier with maintaining the epithelium in order to promote bacterial colonization. These complex strategies represent a new paradigm of bacterial pathogenesis.
Collapse
Affiliation(s)
- Minsoo Kim
- Department of Infectious Disease Control, International Research Center for Infectious Diseases, University of Tokyo 4-6-1, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Enteroaggregative Escherichia coli (EAEC) is responsible for inflammatory diarrhea in diverse populations, but its mechanisms of pathogenesis have not been fully elucidated. We have used a previously characterized polarized intestinal T84 cell model to investigate the effects of infection with EAEC strain 042 on tight junction integrity. We find that infection with strain 042 induces a decrease in transepithelial electrical resistance (TER) compared to uninfected controls and to cells infected with commensal E. coli strain HS. When the infection was limited after 3 h by washing and application of gentamicin, we observed that the TER of EAEC-infected monolayers continued to decline, and they remained low even as long as 48 h after the infection. Cells infected with the afimbrial mutant strain 042aafA exhibited TER measurements similar to those seen in uninfected monolayers, implicating the aggregative adherence fimbriae II (AAF/II) as necessary for barrier dysfunction. Infection with wild-type strain 042 induced aberrant localization of the tight junction proteins claudin-1 and, to a lesser degree, occludin. EAEC-infected T84 cells exhibited irregular shapes, and some cells became elongated and/or enlarged; these effects were not observed after infection with commensal E. coli strain HS or 042aafA. The effects on tight junctions were also observed with AAF/I-producing strain JM221, and an afimbrial mutant was similarly deficient in inducing barrier dysfunction. Our results show that EAEC induces epithelial barrier dysfunction in vitro and implicates the AAF adhesins in this phenotype.
Collapse
|
45
|
Winter SE, Keestra AM, Tsolis RM, Bäumler AJ. The blessings and curses of intestinal inflammation. Cell Host Microbe 2010; 8:36-43. [PMID: 20638640 PMCID: PMC2918243 DOI: 10.1016/j.chom.2010.06.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 04/10/2010] [Accepted: 06/07/2010] [Indexed: 12/19/2022]
Abstract
The intestinal immune system has to strike a delicate balance between initiating inflammatory responses against invading bacterial pathogens and avoiding their induction against microbiota colonizing the lumen. Adequate inflammatory responses against bacterial invasion result in the lumenal secretion of antimicrobial peptides, as well as the release of cytokines in tissue that recruit and activate phagocytes. However, pathogens have evolved to utilize these environmental changes in the inflamed intestine to promote colonization. This review focuses on the costs and benefits of intestinal inflammation and the fine interplay between the host, its microbiota, and enteric pathogens.
Collapse
Affiliation(s)
- Sebastian E. Winter
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| | - A. Marijke Keestra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| |
Collapse
|
46
|
The impact of the microbiota on the pathogenesis of IBD: lessons from mouse infection models. Nat Rev Microbiol 2010; 8:564-77. [PMID: 20622892 DOI: 10.1038/nrmicro2403] [Citation(s) in RCA: 282] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a major human health problem. The bacteria that live in the gut play an important part in the pathogenesis of IBD. However, owing to the complexity of the gut microbiota, our understanding of the roles of commensal and pathogenic bacteria in establishing a healthy intestinal barrier and in its disruption is evolving only slowly. In recent years, mouse models of intestinal inflammatory disorders based on defined bacterial infections have been used intensively to dissect the roles of individual bacterial species and specific bacterial components in the pathogenesis of IBD. In this Review, we focus on the impact of pathogenic and commensal bacteria on IBD-like pathogenesis in mouse infection models and summarize important recent developments.
Collapse
|
47
|
Kang YJ, Otsuka M, van den Berg A, Hong L, Huang Z, Wu X, Zhang DW, Vallance BA, Tobias PS, Han J. Epithelial p38alpha controls immune cell recruitment in the colonic mucosa. PLoS Pathog 2010; 6:e1000934. [PMID: 20532209 PMCID: PMC2880565 DOI: 10.1371/journal.ppat.1000934] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 05/03/2010] [Indexed: 11/19/2022] Open
Abstract
Intestinal epithelial cells (IECs) compose the first barrier against microorganisms in the gastrointestinal tract. Although the NF-kappaB pathway in IECs was recently shown to be essential for epithelial integrity and intestinal immune homeostasis, the roles of other inflammatory signaling pathways in immune responses in IECs are still largely unknown. Here we show that p38alpha in IECs is critical for chemokine expression, subsequent immune cell recruitment into the intestinal mucosa, and clearance of the infected pathogen. Mice with p38alpha deletion in IECs suffer from a sustained bacterial burden after inoculation with Citrobacter rodentium. These animals are normal in epithelial integrity and immune cell function, but fail to recruit CD4(+) T cells into colonic mucosal lesions. The expression of chemokines in IECs is impaired, which appears to be responsible for the impaired T cell recruitment. Thus, p38alpha in IECs contributes to the host immune responses against enteric bacteria by the recruitment of immune cells.
Collapse
Affiliation(s)
- Young Jun Kang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Motoyuki Otsuka
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Arjen van den Berg
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Lixin Hong
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe Huang
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiurong Wu
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Duan-Wu Zhang
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Bruce A. Vallance
- Division of Gastroenterology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Peter S. Tobias
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jiahuai Han
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
48
|
Li Q, Zhang Q, Wang C, Liu X, Qu L, Gu L, Li N, Li J. Altered distribution of tight junction proteins after intestinal ischaemia/reperfusion injury in rats. J Cell Mol Med 2009; 13:4061-76. [PMID: 19929946 PMCID: PMC4516553 DOI: 10.1111/j.1582-4934.2009.00975.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Tight junction (TJ) disruptions have been demonstrated both in vitro and more recently in vivo in infection. However, the molecular basis for changes of TJ during ischaemia-reperfusion (I/R) injury is poorly understood. In the present study, intestinal damage was induced by I/R in an animal model. As assessed by TUNEL and propidium iodide uptake, we showed that I/R injury induced apoptosis as well as necrosis in rat colon, and the frequency of apoptotic and necrotic cells reached the maximum at 5 hrs of reperfusion. Immunofluorescence microscopy revealed that claudins 1, 3 and 5 are strongly expressed in the surface epithelial cells of the colon; however, labelling of all three proteins was present diffusely within cells and no longer focused at the lateral cell boundaries after I/R. Using Western blot analysis, we found that distribution of TJ proteins in membrane microdomains of TJ was markedly affected in I/R injury rats. Occludin, ZO-1, claudin-1 and claudin-3 were completely displaced from TX-100 insoluble fractions to TX-100 soluble fractions, and claudin-5 was partly displaced. The distribution of lipid raft marker protein caveolin-1 was also changed after I/R. I/R injury results in the disruption of TJs, which characterized by relocalization of the claudins 1, 3 and 5 and an increase in intestinal permeability using molecular tracer measurement. I/R injury altered distribution of TJ proteins in vivo that was associated with functional TJ deficiencies.
Collapse
Affiliation(s)
- Qiurong Li
- Institute of General Surgery, Jinling Hospital, Nanjing, China.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Conlin VS, Wu X, Nguyen C, Dai C, Vallance BA, Buchan AMJ, Boyer L, Jacobson K. Vasoactive intestinal peptide ameliorates intestinal barrier disruption associated with Citrobacter rodentium-induced colitis. Am J Physiol Gastrointest Liver Physiol 2009; 297:G735-50. [PMID: 19661153 DOI: 10.1152/ajpgi.90551.2008] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Attaching and effacing bacterial pathogens attach to the apical surface of epithelial cells and disrupt epithelial barrier function, increasing permeability and allowing luminal contents access to the underlying milieu. Previous in vitro studies demonstrated that the neuropeptide vasoactive intestinal peptide (VIP) regulates epithelial paracellular permeability, and the high concentrations and close proximity of VIP-containing nerve fibers to intestinal epithelial cells would support such a function in vivo. The aim of this study was to examine whether VIP treatment modulated Citrobacter rodentium-induced disruption of intestinal barrier integrity and to identify potential mechanisms of action. Administration of VIP had no effect on bacterial attachment although histopathological scoring demonstrated a VIP-induced amelioration of colitis-induced epithelial damage compared with controls. VIP treatment prevented the infection-induced increase in mannitol flux a measure of paracellular permeability, resulting in levels similar to control mice, and immunohistochemical studies demonstrated that VIP prevented the translocation of tight junction proteins: zonula occludens-1, occludin, and claudin-3. Enteropathogenic Escherichia coli (EPEC) infection of Caco-2 monolayers confirmed a protective role for VIP on epithelial barrier function. VIP prevented EPEC-induced increase in long myosin light chain kinase (MLCK) expression and myosin light chain phosphorylation (p-MLC). Furthermore, MLCK inhibition significantly attenuated bacterial-induced epithelial damage both in vivo and in vitro. In conclusion, our results indicate that VIP protects the colonic epithelial barrier by minimizing bacterial-induced redistribution of tight junction proteins in part through actions on MLCK and MLC phosphorylation.
Collapse
Affiliation(s)
- V S Conlin
- Division of Gastroenterology, Child and Family Research Institute, BC Children's Hospital, Vancouver, BC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Lapointe TK, O'Connor PM, Buret AG. The role of epithelial malfunction in the pathogenesis of enteropathogenic E. coli-induced diarrhea. J Transl Med 2009; 89:964-70. [PMID: 19620958 DOI: 10.1038/labinvest.2009.69] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The homeostatic balance of the gastrointestinal tract relies on a single layer of epithelial cells, which assumes both digestive and protective functions. Enteric pathogens, including enteropathogenic Escherichia coli (EPEC), have evolved numerous mechanisms to disrupt basic intestinal epithelial functions, promoting the development of gastrointestinal disorders. Despite its non-invasive nature, EPEC inflicts severe damage to the intestinal mucosa, including the dysregulation of water and solute transport and the disruption of epithelial barrier structure and function. Despite the high prevalence and morbidity of disease caused by EPEC infections, the etiology of its pathogenesis remains incompletely understood. This review integrates the newest findings on EPEC-epithelial interactions with established mechanisms of disease in an attempt to give a comprehensive understanding of the cellular processes whereby this common pathogen may cause diarrheal illness.
Collapse
Affiliation(s)
- Tamia K Lapointe
- Department of Biological Sciences and Inflammation Research Network, University of Calgary, Calgary, AB, Canada
| | | | | |
Collapse
|