1
|
Peterson ST, Dailey KG, Hullahalli K, Sorobetea D, Matsuda R, Sewell J, Yost W, Neill RO, Bobba S, Apenes N, Sherman ME, Balazs GI, Assenmacher CA, Cox A, Lanza M, Shin S, Waldor MK, Brodsky IE. TNF signaling maintains local restriction of bacterial founder populations in intestinal and systemic sites during oral Yersinia infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.639286. [PMID: 40060595 PMCID: PMC11888380 DOI: 10.1101/2025.02.26.639286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Enteroinvasive bacterial pathogens are responsible for an enormous worldwide disease burden that critically affects the young and immunocompromised. Yersinia pseudotuberculosis is a Gram-negative enteric pathogen, closely related to the plague agent Y. pestis, that colonizes intestinal tissues, induces the formation of pyogranulomas along the intestinal tract, and disseminates to systemic organs following oral infection of experimental rodents. Prior studies proposed that systemic tissues were colonized by a pool of intestinal replicating bacteria distinct from populations within Peyer's patches and mesenteric lymph nodes. Whether bacteria within intestinal pyogranulomas serve as the source for systemic dissemination, and the relationship between bacterial populations within different tissue sites is poorly defined. Moreover, the factors that regulate Yersinia colonization and dissemination are not well understood. Here, we demonstrate, using Sequence Tag-based Analysis of Microbial Populations in R (STAMPR), that remarkably small founder populations independently colonize intestinal and systemic tissues. Notably, intestinal pyogranulomas contain clonal populations of bacteria that are restricted and do not spread to other tissues. However, populations of Yersinia are shared among systemic organs and the blood, suggesting that systemic dissemination occurs via hematogenous spread. Finally, we demonstrate that TNF signaling is a key contributor to the bottlenecks limiting both tissue colonization and lymphatic dissemination of intestinal bacterial populations. Altogether, this study reveals previously undescribed aspects of infection dynamics of enteric bacterial pathogens.
Collapse
Affiliation(s)
- Stefan T Peterson
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Katherine G Dailey
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, USA
- Department of Microbiology, Harvard Medical School, Boston, USA
| | - Karthik Hullahalli
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, USA
- Department of Microbiology, Harvard Medical School, Boston, USA
| | - Daniel Sorobetea
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Rina Matsuda
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Jaydeen Sewell
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Winslow Yost
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Rosemary O' Neill
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Suhas Bobba
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolai Apenes
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Matthew E Sherman
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - George I Balazs
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, USA
- Department of Microbiology, Harvard Medical School, Boston, USA
| | | | - Arin Cox
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Matthew Lanza
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston, USA
- Department of Microbiology, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Igor E Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
2
|
Schreiner J, Brettner FEB, Gier S, Vogel-Kindgen S, Windbergs M. Unlocking the potential of microfold cells for enhanced permeation of nanocarriers in oral drug delivery. Eur J Pharm Biopharm 2024; 202:114408. [PMID: 39004319 DOI: 10.1016/j.ejpb.2024.114408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The therapeutic effects of orally administered nanocarriers depend on their ability to effectively permeate the intestinal mucosa, which is one of the major challenges in oral drug delivery. Microfold cells are specialized enterocytes in the intestinal epithelium known for their high transcytosis abilities. This study aimed to compare and evaluate two targeting approaches using surface modifications of polymer-based nanocarriers, whereas one generally addresses enterocytes, and one is directed explicitly to microfold cells via targeting the sialyl LewisA motif on their surface. We characterized the resulting carriers in terms of size and charge, supplemented by scanning electron microscopy to confirm their structural properties. For predictive biological testing and to assess the intended targeting effect, we implemented two human intestinal in vitro models containing microfold-like cells. Both models were thoroughly characterized prior to permeation studies with the different nanocarriers. Our results demonstrated improved transport for both targeted formulations compared to undecorated carriers in the in vitro models. Notably, there was an enhanced uptake in the presence of microfold-like cells, particularly for the nanocarriers directed by the anti-sialyl LewisA antibody. These findings highlight the potential of microfold cell targeting to improve oral administration of drugs and emphasize the importance of using suitable and well-characterized in vitro models for testing novel drug delivery strategies.
Collapse
Affiliation(s)
- Jonas Schreiner
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Felix E B Brettner
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Stefanie Gier
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Sarah Vogel-Kindgen
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
3
|
Fang X, Kang L, Qiu YF, Li ZS, Bai Y. Yersinia enterocolitica in Crohn’s disease. Front Cell Infect Microbiol 2023; 13:1129996. [PMID: 36968108 PMCID: PMC10031030 DOI: 10.3389/fcimb.2023.1129996] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Increasing attention is being paid to the unique roles gut microbes play in both physiological and pathological processes. Crohn’s disease (CD) is a chronic, relapsing, inflammatory disease of the gastrointestinal tract with unknown etiology. Currently, gastrointestinal infection has been proposed as one initiating factor of CD. Yersinia enterocolitica, a zoonotic pathogen that exists widely in nature, is one of the most common bacteria causing acute infectious gastroenteritis, which displays clinical manifestations similar to CD. However, the specific role of Y. enterocolitica in CD is controversial. In this Review, we discuss the current knowledge on how Y. enterocolitica and derived microbial compounds may link to the pathogenesis of CD. We highlight examples of Y. enterocolitica-targeted interventions in the diagnosis and treatment of CD, and provide perspectives for future basic and translational investigations on this topic.
Collapse
Affiliation(s)
| | | | | | | | - Yu Bai
- *Correspondence: Zhao-Shen Li, ; Yu Bai,
| |
Collapse
|
4
|
Abstract
Pathogen attachment to host tissue is critical in the progress of many infections. Bacteria use adhesion in vivo to stabilize colonization and subsequently regulate the deployment of contact-dependent virulence traits. To specifically target host cells, they decorate themselves with adhesins, proteins that bind to mammalian cell surface receptors. One common assumption is that adhesin-receptor interactions entirely govern bacterial attachment. However, how adhesins engage with their receptors in an in vivo-like context remains unclear, in particular under the influence of a heterogeneous mechanical microenvironment. We here investigate the biophysical processes governing bacterial adhesion to host cells using a tunable adhesin-receptor system. By dynamically visualizing attachment, we found that bacterial adhesion to host cell surface, unlike adhesion to inert surfaces, involves two consecutive steps. Bacteria initially attach to their host without engaging adhesins. This step lasts about 1 min, during which bacteria can easily detach. We found that at this stage, the glycocalyx, a layer of glycosylated proteins and lipids, shields the host cell by keeping adhesins away from their receptor ligand. In a second step, adhesins engage with their target receptors to strengthen attachment for minutes to hours. The active properties of the membrane, endowed by the actin cytoskeleton, strengthen specific adhesion. Altogether, our results demonstrate that adhesin-ligand binding is not the sole regulator of bacterial adhesion. In fact, the host cell’s surface mechanical microenvironment mediates the physical interactions between host and bacteria, thereby playing an essential role in the onset of infection.
Collapse
|
5
|
Khalid DM, Abbas BA. Prevalence, antibiotic susceptibility, and virulence factors of Yersinia enterocolitica isolated from raw milk in Basrah, Iraq. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2021. [DOI: 10.15547/bjvm.2019-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A total of one hundred and fifty cow, buffalo, and sheep milk samples were collected from several markets in Basrah, Iraq (50 samples from each species). All milk samples were subjected to enrichment in TSB and cold enrichment in PBS, then cultured on YSA agar in order to obtain Yersinia species. The highest growth obtained by TSB enrichment was seen in cow milk (24%) followed by buffalo milk (22%) and sheep milk (12%). The results of PBS enrichment showed the highest growth in cow milk (14%) followed by buffalo (8%) and sheep milk (2%). The results showed that both cow and buffalo milk samples were contaminated by Y. enterocolitica at 8% while the prevalence in sheep milk was 4%. Ten isolates from different sources in the current study were examined for their susceptibility to 10 antibiotics. The highest susceptibility (100%) was found towards streptomycin, azithromycin and gentamicin, followed by ciprofloxacin and chloramphenicol, 93.3% for each. Low susceptibility was found toward vancomycin (6.66%) and cloxacillin (33.3%). 16S rRNA sequencing showed homology with previously annotated strains at GenBank of National Centre for Biotechnology (NCBI). Multiple sequence alignments exhibited one difference between the sequences at the locus 764. The phylogenic tree of the results demonstrated that the local isolates were closely related to strains previously reported from China. All Yersinia enterocolitica strains had the inv gene. In contrast, the ail gene was found in one strain (10%) while the yad gene appeared in 50% of the investigated strains.
Collapse
|
6
|
Whelan R, McVicker G, Leo JC. Staying out or Going in? The Interplay between Type 3 and Type 5 Secretion Systems in Adhesion and Invasion of Enterobacterial Pathogens. Int J Mol Sci 2020; 21:E4102. [PMID: 32521829 PMCID: PMC7312957 DOI: 10.3390/ijms21114102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Enteric pathogens rely on a variety of toxins, adhesins and other virulence factors to cause infections. Some of the best studied pathogens belong to the Enterobacterales order; these include enteropathogenic and enterohemorrhagic Escherichia coli, Shigella spp., and the enteropathogenic Yersiniae. The pathogenesis of these organisms involves two different secretion systems, a type 3 secretion system (T3SS) and type 5 secretion systems (T5SSs). The T3SS forms a syringe-like structure spanning both bacterial membranes and the host cell plasma membrane that translocates toxic effector proteins into the cytoplasm of the host cell. T5SSs are also known as autotransporters, and they export part of their own polypeptide to the bacterial cell surface where it exerts its function, such as adhesion to host cell receptors. During infection with these enteropathogens, the T3SS and T5SS act in concert to bring about rearrangements of the host cell cytoskeleton, either to invade the cell, confer intracellular motility, evade phagocytosis or produce novel structures to shelter the bacteria. Thus, in these bacteria, not only the T3SS effectors but also T5SS proteins could be considered "cytoskeletoxins" that bring about profound alterations in host cell cytoskeletal dynamics and lead to pathogenic outcomes.
Collapse
Affiliation(s)
| | | | - Jack C. Leo
- Antimicrobial Resistance, Omics and Microbiota Group, Department of Biosciences, Nottingham Trent University, Nottingham NG1 4FQ, UK; (R.W.); (G.M.)
| |
Collapse
|
7
|
Dillon A, Lo DD. M Cells: Intelligent Engineering of Mucosal Immune Surveillance. Front Immunol 2019; 10:1499. [PMID: 31312204 PMCID: PMC6614372 DOI: 10.3389/fimmu.2019.01499] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
M cells are specialized intestinal epithelial cells that provide the main machinery for sampling luminal microbes for mucosal immune surveillance. M cells are usually found in the epithelium overlying organized mucosal lymphoid tissues, but studies have identified multiple distinct lineages of M cells that are produced under different conditions, including intestinal inflammation. Among these lineages there is a common morphology that helps explain the efficiency of M cells in capturing luminal bacteria and viruses; in addition, M cells recruit novel cellular mechanisms to transport the particles across the mucosal barrier into the lamina propria, a process known as transcytosis. These specializations used by M cells point to a novel engineering of cellular machinery to selectively capture and transport microbial particles of interest. Because of the ability of M cells to effectively violate the mucosal barrier, the circumstances of M cell induction have important consequences. Normal immune surveillance insures that transcytosed bacteria are captured by underlying myeloid/dendritic cells; in contrast, inflammation can induce development of new M cells not accompanied by organized lymphoid tissues, resulting in bacterial transcytosis with the potential to amplify inflammatory disease. In this review, we will discuss our own perspectives on the life history of M cells and also raise a few questions regarding unique aspects of their biology among epithelia.
Collapse
Affiliation(s)
- Andrea Dillon
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
8
|
Basmaciyan L, Bon F, Paradis T, Lapaquette P, Dalle F. " Candida Albicans Interactions With The Host: Crossing The Intestinal Epithelial Barrier". Tissue Barriers 2019; 7:1612661. [PMID: 31189436 PMCID: PMC6619947 DOI: 10.1080/21688370.2019.1612661] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023] Open
Abstract
Formerly a commensal organism of the mucosal surfaces of most healthy individuals, Candida albicans is an opportunistic pathogen that causes infections ranging from superficial to the more life-threatening disseminated infections, especially in the ever-growing population of vulnerable patients in the hospital setting. In these situations, the fungus takes advantage of its host following a disturbance in the host defense system and/or the mucosal microbiota. Overwhelming evidence suggests that the gastrointestinal tract is the main source of disseminated C. albicans infections. Major risk factors for disseminated candidiasis include damage to the mucosal intestinal barrier, immune dysfunction, and dysbiosis of the resident microbiota. A better understanding of C. albicans' interaction with the intestinal epithelial barrier will be useful for designing future therapies to avoid systemic candidiasis. In this review, we provide an overview of the current knowledge regarding the mechanisms of pathogenicity that allow the fungus to reach and translocate the gut barrier.
Collapse
Affiliation(s)
- Louise Basmaciyan
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Fabienne Bon
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Tracy Paradis
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Pierre Lapaquette
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| | - Frédéric Dalle
- Laboratoire de Parasitologie-Mycologie, Plateforme de Biologie Hospitalo-Universitaire Gérard Mack, Dijon France
- UMR PAM Univ Bourgogne Franche-Comté - AgroSup Dijon - Equipe Vin, Aliment, Microbiologie, Stress, Dijon, France
| |
Collapse
|
9
|
Meuskens I, Saragliadis A, Leo JC, Linke D. Type V Secretion Systems: An Overview of Passenger Domain Functions. Front Microbiol 2019; 10:1163. [PMID: 31214135 PMCID: PMC6555100 DOI: 10.3389/fmicb.2019.01163] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022] Open
Abstract
Bacteria secrete proteins for different purposes such as communication, virulence functions, adhesion to surfaces, nutrient acquisition, or growth inhibition of competing bacteria. For secretion of proteins, Gram-negative bacteria have evolved different secretion systems, classified as secretion systems I through IX to date. While some of these systems consist of multiple proteins building a complex spanning the cell envelope, the type V secretion system, the subject of this review, is rather minimal. Proteins of the Type V secretion system are often called autotransporters (ATs). In the simplest case, a type V secretion system consists of only one polypeptide chain with a β-barrel translocator domain in the membrane, and an extracellular passenger or effector region. Depending on the exact domain architecture of the protein, type V secretion systems can be further separated into sub-groups termed type Va through e, and possibly another recently identified subtype termed Vf. While this classification works well when it comes to the architecture of the proteins, this is not the case for the function(s) of the secreted passenger. In this review, we will give an overview of the functions of the passengers of the different AT classes, shedding more light on the variety of functions carried out by type V secretion systems.
Collapse
Affiliation(s)
| | | | | | - Dirk Linke
- Department of Biosciences, Section for Genetics and Evolutionary Biology, University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Walker D, Gibbons J, Harris JD, Taylor CS, Scott C, Paterson GK, Morrison LR. Systemic Yersinia pseudotuberculosis as a Cause of Osteomyelitis in a Captive Ring-tailed Lemur (Lemur catta). J Comp Pathol 2018; 164:27-31. [PMID: 30360909 DOI: 10.1016/j.jcpa.2018.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 11/29/2022]
Abstract
Yersinia pseudotuberculosis and Yersinia enterocolitica are ubiquitous pathogens with wildlife and domestic animal reservoirs. Outbreaks of 'non-plague' yersiniosis in man and non-human primates are reported frequently (including zoological specimens and research breeding colonies) and are usually characterized by enteritis, mesenteric lymphadenitis and occasionally organ abscessation. In people, non-septic reactive arthritis is a common sequela to yersiniosis. However, there have been rare reports in people of septic arthritis and osteomyelitis because of active systemic infection with Y. pseudotuberculosis. Osteomyelitis has also been reported rarely in historical yersiniosis outbreaks in farmed turkeys in England and the USA. This paper reports the first case of osteomyelitis caused by systemic infection with Y. pseudotuberculosis O:1 in a non-human primate, a captive ring-tailed lemur (Lemur catta). The lemur had a short clinical history of hyporexia and weight loss with reduction in mobility, especially of the left hindlimb. On post-mortem examination there was evidence of multi-organ abscessation. In addition, severe necrosis, inflammation and large bacterial colonies were present in the musculature, periosteum and bone marrow in the hip, ribs and a vertebra at the cervicothoracic junction. Osteomyelitis should be considered as a rare clinical presentation in non-human primates with systemic Y. pseudotuberculosis infection.
Collapse
Affiliation(s)
- D Walker
- Royal (Dick) School of Veterinary Studies, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, UK.
| | - J Gibbons
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - J D Harris
- Royal (Dick) School of Veterinary Studies, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, UK
| | - C S Taylor
- Royal (Dick) School of Veterinary Studies, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, UK
| | - C Scott
- Struthers and Scott Veterinary Practice, Doune, Perthshire FK16 6EH, UK
| | - G K Paterson
- Royal (Dick) School of Veterinary Studies, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, UK
| | - L R Morrison
- Royal (Dick) School of Veterinary Studies, The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, UK
| |
Collapse
|
11
|
Beyond the Matrix: The Many Non-ECM Ligands for Integrins. Int J Mol Sci 2018; 19:ijms19020449. [PMID: 29393909 PMCID: PMC5855671 DOI: 10.3390/ijms19020449] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/21/2018] [Accepted: 01/30/2018] [Indexed: 12/17/2022] Open
Abstract
The traditional view of integrins portrays these highly conserved cell surface receptors as mediators of cellular attachment to the extracellular matrix (ECM), and to a lesser degree, as coordinators of leukocyte adhesion to the endothelium. These canonical activities are indispensable; however, there is also a wide variety of integrin functions mediated by non-ECM ligands that transcend the traditional roles of integrins. Some of these unorthodox roles involve cell-cell interactions and are engaged to support immune functions such as leukocyte transmigration, recognition of opsonization factors, and stimulation of neutrophil extracellular traps. Other cell-cell interactions mediated by integrins include hematopoietic stem cell and tumor cell homing to target tissues. Integrins also serve as cell-surface receptors for various growth factors, hormones, and small molecules. Interestingly, integrins have also been exploited by a wide variety of organisms including viruses and bacteria to support infectious activities such as cellular adhesion and/or cellular internalization. Additionally, the disruption of integrin function through the use of soluble integrin ligands is a common strategy adopted by several parasites in order to inhibit blood clotting during hematophagy, or by venomous snakes to kill prey. In this review, we strive to go beyond the matrix and summarize non-ECM ligands that interact with integrins in order to highlight these non-traditional functions of integrins.
Collapse
|
12
|
Vigilance or Subversion? Constitutive and Inducible M Cells in Mucosal Tissues. Trends Immunol 2017; 39:185-195. [PMID: 28958392 DOI: 10.1016/j.it.2017.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 02/08/2023]
Abstract
Microfold (M) cells are epithelial cells present in mucosal tissues and specialized for the capture of luminal microparticles and their delivery to underlying immune cells; thus, they are crucial participants in mucosal immune surveillance. Multiple phenotypic subsets of M cells have now been described, all sharing a unique apical morphology that provides clues to their ability to capture microbial particles. The existence of diverse M cell phenotypes, especially inflammation-inducible M cells, provides an intriguing puzzle: some variants may augment luminal surveillance to boost mucosal immunity, while others may promote microbial access to tissues. Here, I consider the unique induction requirements of each M cell subset and functional differences, highlighting the potentially distinct consequences in mucosal immunity.
Collapse
|
13
|
Jariwala R, Mandal H, Bagchi T. Indigenous lactobacilli strains of food and human sources reverse enteropathogenic E. coli O26:H11-induced damage in intestinal epithelial cell lines: effect on redistribution of tight junction proteins. MICROBIOLOGY-SGM 2017; 163:1263-1272. [PMID: 28771130 DOI: 10.1099/mic.0.000507] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The aim of the study was to investigate the neutralizing effect of lactobacilli isolated from indigenous food and human sources on enteropathogenic Escherichia coli (EPEC) O26 : H11-induced epithelial barrier dysfunction in vitro. This was assessed by transepithelial electrical resistance (TEER) and permeability assays using intestinal cell lines, HT-29 and Caco-2. Furthermore, the expression and distribution of tight junction (TJ) proteins were analysed by qRT-PCR and immunofluorescence assay, respectively. The nine strains used in the study were from different species viz. Lactobacillus fermentum, Lactobacillushelveticus, Lactobacillus salivarius and Lactobacillus plantarum. All strains were able to reverse the decrease in TEER and corresponding increase in permeability across E. coli-infected monolayers. Maximum reversal was observed after 18 h [up to 93.8±2.0 % by L. rhamnosus GG followed by L. fermentum IIs11.2 (92.6±2.2 %) and L. plantarum GRI-2 (91.9±0.9 %)] of lactobacilli exposure following EPEC O26 : H11 infection. All strains were able to redistribute the TJ proteins to the cell periphery either partially or completely. Moreover, L. helveticus FA-7 was also able to significantly increase the mRNA expression of ZO-1 and claudin-1 (2.5-fold and 3.0-fold, respectively; P<0.05). The rapid reversal observed by these strains could be mostly because of the redistribution rather than increased mRNA expression of TJ proteins. In conclusion, L. helveticus FA-7, L. fermentum FA-1 and L. plantarum GRI-2 were good in all the aspects studied, and the other strains were good in some aspects. L. helveticus FA-7, L. fermentum FA-1 and L. plantarum GRI-2 can therefore be used for potential therapeutic purpose against intestinal epithelial dysfunction.
Collapse
Affiliation(s)
- Ruchi Jariwala
- Department of Microbiology and Biotechnology Centre, Faculty of Science, M. S. University of Baroda, Vadodara 390 002, Gujarat, India
| | - Hemanti Mandal
- Department of Microbiology and Biotechnology Centre, Faculty of Science, M. S. University of Baroda, Vadodara 390 002, Gujarat, India
| | - Tamishraha Bagchi
- Department of Microbiology and Biotechnology Centre, Faculty of Science, M. S. University of Baroda, Vadodara 390 002, Gujarat, India.,Present address: Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| |
Collapse
|
14
|
Innovative Solutions to Sticky Situations: Antiadhesive Strategies for Treating Bacterial Infections. Microbiol Spectr 2017; 4. [PMID: 27227305 DOI: 10.1128/microbiolspec.vmbf-0023-2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bacterial adherence to host tissue is an essential process in pathogenesis, necessary for invasion and colonization and often required for the efficient delivery of toxins and other bacterial effectors. As existing treatment options for common bacterial infections dwindle, we find ourselves rapidly approaching a tipping point in our confrontation with antibiotic-resistant strains and in desperate need of new treatment options. Bacterial strains defective in adherence are typically avirulent and unable to cause infection in animal models. The importance of this initial binding event in the pathogenic cascade highlights its potential as a novel therapeutic target. This article seeks to highlight a variety of strategies being employed to treat and prevent infection by targeting the mechanisms of bacterial adhesion. Advancements in this area include the development of novel antivirulence therapies using small molecules, vaccines, and peptides to target a variety of bacterial infections. These therapies target bacterial adhesion through a number of mechanisms, including inhibition of pathogen receptor biogenesis, competition-based strategies with receptor and adhesin analogs, and the inhibition of binding through neutralizing antibodies. While this article is not an exhaustive description of every advancement in the field, we hope it will highlight several promising examples of the therapeutic potential of antiadhesive strategies.
Collapse
|
15
|
Parnell EA, Walch EM, Lo DD. Inducible Colonic M Cells Are Dependent on TNFR2 but Not Ltβr, Identifying Distinct Signalling Requirements for Constitutive Versus Inducible M Cells. J Crohns Colitis 2017; 11:751-760. [PMID: 27932454 PMCID: PMC5881705 DOI: 10.1093/ecco-jcc/jjw212] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/16/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS M cells associated with organised lymphoid tissues such as intestinal Peyer's patches provide surveillance of the intestinal lumen. Inflammation or infection in the colon can induce an M cell population associated with lymphoid infiltrates; paradoxically, induction is dependent on the inflammatory cytokine tumour necrosis factor [TNF]-α. Anti-TNFα blockade is an important therapeutic in inflammatory bowel disease, so understanding the effects of TNFα signalling is important in refining therapeutics. METHODS To dissect pro-inflammatory signals from M cell inductive signals, we used confocal microscopy image analysis to assess requirements for specific cytokine receptor signals using TNF receptor 1 [TNFR1] and 2 [TNFR2] knockouts [ko] back-crossed to the PGRP-S-dsRed transgene; separate groups were treated with soluble lymphotoxin β receptor [sLTβR] to block LTβR signalling. All groups were treated with dextran sodium sulphate [DSS] to induce colitis. RESULTS Deficiency of TNFR1 or TNFR2 did not prevent DSS-induced inflammation nor induction of stromal cell expression of receptor activator of nuclear factor kappa-B ligand [RANKL], but absence of TNFR2 prevented M cell induction. LTβR blockade had no effect on M cell induction, but it appeared to reduce RANKL induction below adjacent M cells. CONCLUSIONS TNFR2 is required for inflammation-inducible M cells, indicating that constitutive versus inflammation-inducible M cells depend on different triggers. The inducible M cell dependence on TNFR2 suggests that this specific subset is dependent on TNFα in addition to a presumed requirement for RANKL. Since inducible M cell function will influence immune responses, selective blockade of TNFα may affect colonic inflammation.
Collapse
Affiliation(s)
- Erinn A. Parnell
- Division of Biomedical Sciences, University of California Riverside School of Medicine,Riverside, CA, USA.
| | - Erin M. Walch
- Division of Biomedical Sciences, University of California Riverside School of Medicine,Riverside, CA, USA.
| | - David D. Lo
- Division of Biomedical Sciences, University of California Riverside School of Medicine,Riverside, CA, USA.
| |
Collapse
|
16
|
Role of Host Type IA Phosphoinositide 3-Kinase Pathway Components in Invasin-Mediated Internalization of Yersinia enterocolitica. Infect Immun 2016; 84:1826-1841. [PMID: 27068087 DOI: 10.1128/iai.00142-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/03/2016] [Indexed: 02/07/2023] Open
Abstract
Many bacterial pathogens subvert mammalian type IA phosphoinositide 3-kinase (PI3K) in order to induce their internalization into host cells. How PI3K promotes internalization is not well understood. Also unclear is whether type IA PI3K affects different pathogens through similar or distinct mechanisms. Here, we performed an RNA interference (RNAi)-based screen to identify components of the type IA PI3K pathway involved in invasin-mediated entry of Yersinia enterocolitica, an enteropathogen that causes enteritis and lymphadenitis. The 69 genes targeted encode known upstream regulators or downstream effectors of PI3K. A similar RNAi screen was previously performed with the food-borne bacterium Listeria monocytogenes The results of the screen with Y. enterocolitica indicate that at least nine members of the PI3K pathway are needed for invasin-mediated entry. Several of these proteins, including centaurin-α1, Dock180, focal adhesion kinase (FAK), Grp1, LL5α, LL5β, and PLD2 (phospholipase D2), were recruited to sites of entry. In addition, centaurin-α1, FAK, PLD2, and mTOR were required for remodeling of the actin cytoskeleton during entry. Six of the human proteins affecting invasin-dependent internalization also promote InlB-mediated entry of L. monocytogenes Our results identify several host proteins that mediate invasin-induced effects on the actin cytoskeleton and indicate that a subset of PI3K pathway components promote internalization of both Y. enterocolitica and L. monocytogenes.
Collapse
|
17
|
Mononuclear phagocytes contribute to intestinal invasion and dissemination of Yersinia enterocolitica. Int J Med Microbiol 2016; 306:357-66. [PMID: 27107739 DOI: 10.1016/j.ijmm.2016.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/05/2016] [Accepted: 04/14/2016] [Indexed: 12/30/2022] Open
Abstract
Enteropathogenic Yersinia enterocolitica (Ye) enters the host via contaminated food. After colonisation of the small intestine Ye invades the Peyer's patches (PPs) via M cells and disseminates to the mesenteric lymph nodes (MLNs), spleen and liver. Whether Ye uses other invasion routes and which pathogenicity factors are required remains elusive. Oral infection of lymphotoxin-β-receptor deficient mice lacking PPs and MLNs with Ye revealed similar bacterial load in the spleen 1h post infection as wild-type mice, demonstrating a PP-independent dissemination route for Ye. Immunohistological analysis of the small intestine revealed Ye in close contact with mononuclear phagocytes (MPs), specifically CX3CR1(+) monocyte-derived cells (MCs) as well as CD103(+) dendritic cells (DCs). This finding was confirmed by flow cytometry and imaging flow cytometry analysis of lamina propria (LP) leukocytes showing CD103(+) DCs and MCs with intracellular Ye. Uptake of Ye by LP CD103(+) DCs and MCs was dependent on the pathogenicity factor invasin, whereas the adhesin YadA was dispensable as demonstrated by Ye deletion mutants. Furthermore, Ye were found exclusively associated with CD103(+) DCs in the MLNs from wild-type mice, but not from CCR7(-/-) mice, demonstrating a CCR7 dependent transport of Ye by CD103(+) DCs from LP to the MLNs. In contrast, dissemination of Ye to the spleen was dependent on MCs as significantly less Ye could be recovered from the spleen of CX3CR1(GFP/GFP) mice compared to wild-type mice. Altogether, MCs and CD103(+) DCs contribute to immediate invasion and dissemination of Ye. This together with data from other bacteria suggests MPs as general pathogenic entry site in the intestine.
Collapse
|
18
|
Khan TA, Wang X, Maynard JA. Inclusion of an RGD Motif Alters Invasin Integrin-Binding Affinity and Specificity. Biochemistry 2016; 55:2078-90. [DOI: 10.1021/acs.biochem.5b01243] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tarik A. Khan
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Xianzhe Wang
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Jennifer A. Maynard
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
19
|
Kenngott EE, Kiefer R, Schneider-Daum N, Hamann A, Schneider M, Schmitt MJ, Breinig F. Surface-modified yeast cells: A novel eukaryotic carrier for oral application. J Control Release 2016; 224:1-7. [DOI: 10.1016/j.jconrel.2015.12.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 12/11/2022]
|
20
|
Boyle EC, Dombrowsky H, Sarau J, Braun J, Aepfelbacher M, Lautenschläger I, Grassl GA. Ex vivo perfusion of the isolated rat small intestine as a novel model of Salmonella enteritis. Am J Physiol Gastrointest Liver Physiol 2016; 310:G55-63. [PMID: 26564721 DOI: 10.1152/ajpgi.00444.2014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 11/11/2015] [Indexed: 01/31/2023]
Abstract
Using an ex vivo perfused rat small intestinal model, we examined pathological changes to the tissue, inflammation induction, as well as dynamic changes to smooth muscle activity, metabolic competence, and luminal fluid accumulation during short-term infection with the enteropathogenic bacteria Salmonella enterica serovar Typhimurium and Yersinia enterocolitica. Although few effects were seen upon Yersinia infection, this system accurately modeled key aspects associated with Salmonella enteritis. Our results confirmed the importance of the Salmonella Pathogenicity Island 1 (SPI1)-encoded type 3 secretion system (T3SS) in pathology, tissue invasion, inflammation induction, and fluid secretion. Novel physiological consequences of Salmonella infection of the small intestine were also identified, namely, SPI-1-dependent vasoconstriction and SPI-1-independent reduction in the digestive and absorptive functions of the epithelium. Importantly, this is the first small animal model that allows for the study of Salmonella-induced fluid secretion. Another major advantage of this model is that one can specifically determine the contribution of resident cell populations. Accordingly, we can conclude that recruited cell populations were not involved in the pathological damage, inflammation induction, fluid accumulation, nutrient absorption deficiency, and vasoconstriction observed. Although fluid loss induced by Salmonella infection is hypothesized to be due to damage caused by recruited neutrophils, our data suggest that bacterial invasion and inflammation induction in resident cell populations are sufficient for fluid loss into the lumen. In summary, this model is a novel and useful tool that allows for detailed examination of the early physiopathological effects of Salmonella infection on the small intestine.
Collapse
Affiliation(s)
- Erin C Boyle
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Bernhard Nocht Institute of Tropical Medicine, Hamburg, Germany
| | - Heike Dombrowsky
- Priority Area Asthma and Allergy, Research Center Borstel, Borstel, Germany
| | - Jürgen Sarau
- Priority Area Asthma and Allergy, Research Center Borstel, Borstel, Germany
| | - Janin Braun
- Priority Area Infections, Models of Inflammation, Research Center Borstel, Borstel, Germany; Institute for Experimental Medicine, Christian-Albrechts University Kiel, Kiel, Germany
| | - Martin Aepfelbacher
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingmar Lautenschläger
- Priority Area Asthma and Allergy, Research Center Borstel, Borstel, Germany; Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany; and
| | - Guntram A Grassl
- Priority Area Infections, Models of Inflammation, Research Center Borstel, Borstel, Germany; Institute for Experimental Medicine, Christian-Albrechts University Kiel, Kiel, Germany; Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| |
Collapse
|
21
|
Li H, Ning P, Lin Z, Liang W, Kang K, He L, Zhang Y. Co-expression of the C-terminal domain of Yersinia enterocolitica invasin enhances the efficacy of classical swine-fever-vectored vaccine based on human adenovirus. J Biosci 2015; 40:79-90. [PMID: 25740144 DOI: 10.1007/s12038-014-9495-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The use of adenovirus vector-based vaccines is a promising approach for generating antigen-specific immune responses. Improving vaccine potency is necessary in other approaches to address their inadequate protection for the majority of infectious diseases. This study is the first to reconstruct a recombinant replication-defective human adenovirus co-expressing E2 and invasin C-terminal (InvC) glycoproteins (rAd-E2-InvC). rAd-E2-InvC with 2 x 10(6) TCID50 was intramuscularly administered two times to CSFV-free pigs at 14 day intervals. No adverse clinical reactions were observed in any of the pigs after the vaccination. The CSFV E2-specific antibody titer was significantly higher in the rAd-E2-InvC group than that in the rAdV-E2 group as measured by NPLA and blocking ELISA. Pigs immunized with rAd-E2-InvC were completely protected against lethal challenge. Neither CSFV RNA nor pathological changes were detected in the tissues after CSFV challenge. These results demonstrate that rAd-E2-InvC could be an alternative to the existing CSF vaccine. Moreover, InvC that acts as an adjuvant could enhance the immunogenicity of rAdV-E2 and induce high CSFV E2-specific antibody titer and protection level.
Collapse
Affiliation(s)
- Helin Li
- College of Veterinary Medicine, Northwest A and F University, Yangling 712100, Shaanxi, China
| | | | | | | | | | | | | |
Collapse
|
22
|
Albac S, Schmitz A, Lopez-Alayon C, d'Enfert C, Sautour M, Ducreux A, Labruère-Chazal C, Laue M, Holland G, Bonnin A, Dalle F. Candida albicansis able to use M cells as a portal of entry across the intestinal barrierin vitro. Cell Microbiol 2015; 18:195-210. [DOI: 10.1111/cmi.12495] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 07/21/2015] [Indexed: 01/18/2023]
Affiliation(s)
- Sandrine Albac
- UMR 1347, Université de Bourgogne Franche-Comté; 17 Rue Sully, BP 86 510 F-21065 Dijon Cedex France
| | - Antonin Schmitz
- UMR 1347, Université de Bourgogne Franche-Comté; 17 Rue Sully, BP 86 510 F-21065 Dijon Cedex France
| | - Carolina Lopez-Alayon
- UMR 1347, Université de Bourgogne Franche-Comté; 17 Rue Sully, BP 86 510 F-21065 Dijon Cedex France
| | - Christophe d'Enfert
- Institut Pasteur; Unité Biologie et Pathogénicité Fongiques, Département Mycologie; Paris France
- INRA; USC 2019; Paris France
| | - Marc Sautour
- UMR 1347, Université de Bourgogne Franche-Comté; 17 Rue Sully, BP 86 510 F-21065 Dijon Cedex France
- Centre Hospitalier Universitaire; Service de Parasitologie Mycologie; 2 Rue Angélique Ducoudray F-21070 Dijon Cedex France
| | - Amandine Ducreux
- UMR 1347, Université de Bourgogne Franche-Comté; 17 Rue Sully, BP 86 510 F-21065 Dijon Cedex France
| | - Catherine Labruère-Chazal
- Université de Bourgogne Franche-Comté; Institut de Mathématiques de Bourgogne, UFR Sciences et Techniques; Dijon France
| | - Michael Laue
- Robert Koch-Institute; Centre for Biological Threats and Special Pathogens, Advanced Light and Electron Microscopy; Nordufer 20 13353 Berlin Germany
| | - Gudrun Holland
- Robert Koch-Institute; Centre for Biological Threats and Special Pathogens, Advanced Light and Electron Microscopy; Nordufer 20 13353 Berlin Germany
| | - Alain Bonnin
- UMR 1347, Université de Bourgogne Franche-Comté; 17 Rue Sully, BP 86 510 F-21065 Dijon Cedex France
- Centre Hospitalier Universitaire; Service de Parasitologie Mycologie; 2 Rue Angélique Ducoudray F-21070 Dijon Cedex France
| | - Frederic Dalle
- UMR 1347, Université de Bourgogne Franche-Comté; 17 Rue Sully, BP 86 510 F-21065 Dijon Cedex France
- Centre Hospitalier Universitaire; Service de Parasitologie Mycologie; 2 Rue Angélique Ducoudray F-21070 Dijon Cedex France
| |
Collapse
|
23
|
Bringer MA, Darfeuille-Michaud A. Bacterial Adhesion to Intestinal Mucosa. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00048-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
24
|
Heroven AK, Böhme K, Dersch P. The Csr/Rsm system of Yersinia and related pathogens. RNA Biol 2014; 9:379-91. [DOI: 10.4161/rna.19333] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
25
|
Pathogenesis of human enterovirulent bacteria: lessons from cultured, fully differentiated human colon cancer cell lines. Microbiol Mol Biol Rev 2014; 77:380-439. [PMID: 24006470 DOI: 10.1128/mmbr.00064-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hosts are protected from attack by potentially harmful enteric microorganisms, viruses, and parasites by the polarized fully differentiated epithelial cells that make up the epithelium, providing a physical and functional barrier. Enterovirulent bacteria interact with the epithelial polarized cells lining the intestinal barrier, and some invade the cells. A better understanding of the cross talk between enterovirulent bacteria and the polarized intestinal cells has resulted in the identification of essential enterovirulent bacterial structures and virulence gene products playing pivotal roles in pathogenesis. Cultured animal cell lines and cultured human nonintestinal, undifferentiated epithelial cells have been extensively used for understanding the mechanisms by which some human enterovirulent bacteria induce intestinal disorders. Human colon carcinoma cell lines which are able to express in culture the functional and structural characteristics of mature enterocytes and goblet cells have been established, mimicking structurally and functionally an intestinal epithelial barrier. Moreover, Caco-2-derived M-like cells have been established, mimicking the bacterial capture property of M cells of Peyer's patches. This review intends to analyze the cellular and molecular mechanisms of pathogenesis of human enterovirulent bacteria observed in infected cultured human colon carcinoma enterocyte-like HT-29 subpopulations, enterocyte-like Caco-2 and clone cells, the colonic T84 cell line, HT-29 mucus-secreting cell subpopulations, and Caco-2-derived M-like cells, including cell association, cell entry, intracellular lifestyle, structural lesions at the brush border, functional lesions in enterocytes and goblet cells, functional and structural lesions at the junctional domain, and host cellular defense responses.
Collapse
|
26
|
Abstract
Oral vaccines are safe and easy to administer and convenient for all ages. They have been successfully developed to protect from many infectious diseases acquired through oral transmission. We recently found in animal models that formulation of oral vaccines in a nanoparticle-releasing microparticle delivery system is a viable approach for selectively inducing large intestinal protective immunity against infections at rectal and genital mucosae. These large-intestine targeted oral vaccines are a potential substitute for the intracolorectal immunization, which has been found to be effective against rectogenital infections but is not feasible for mass vaccination. Moreover, the newly developed delivery system can be modified to selectively target either the small or large intestine for immunization and accordingly revealed a regionalized immune system in the gut. Future applications and research endeavors suggested by the findings are discussed.
Collapse
Affiliation(s)
- Qing Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Institute of Materia Medica; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing, China,Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study; Institute of Materia Medica; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing, China,Correspondence to: Qing Zhu, and Jay A. Berzofsky,
| | - Jay A. Berzofsky
- Vaccine Branch; National Cancer Institute; National Institutes of Health; Bethesda, MD USA,Correspondence to: Qing Zhu, and Jay A. Berzofsky,
| |
Collapse
|
27
|
Kochut A, Dersch P. Bacterial invasion factors: tools for crossing biological barriers and drug delivery? Eur J Pharm Biopharm 2012. [PMID: 23207324 DOI: 10.1016/j.ejpb.2012.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The oral route is the preferential route of drug delivery in humans. However, effective delivery through the gastrointestinal tract is often hampered by the low permeability of the intestinal epithelium. One possibility to overcome this problem is the encapsulation of drugs inside nanoparticulate systems, containing targeting moieties with cell invasive properties. The bioinvasive features of the delivery system could be provided by the attachment of bacterial invasion factors, which promote efficient uptake into host cells and mediate rapid transcytosis of the pathogen through the intestinal epithelium. This review gives an overview of bacterial invasion systems. The molecular structure and function of suitable bacterial invasins, their relative values as targeting agents and possible pitfalls of their use are described. The potential of bioinvasive drug delivery systems is mainly presented on the basis of the well-characterized Yersinia invasin protein, which enters M cells to gain access to subepithelial layers of the gastrointestinal tract, but alternative approaches and future prospects for oral drug delivery are also discussed.
Collapse
Affiliation(s)
- Annika Kochut
- Department of Molecular Infection Biology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | |
Collapse
|
28
|
Tonry JH, Popov SG, Narayanan A, Kashanchi F, Hakami RM, Carpenter C, Bailey C, Chung MC. In vivo murine and in vitro M-like cell models of gastrointestinal anthrax. Microbes Infect 2012; 15:37-44. [PMID: 23108317 DOI: 10.1016/j.micinf.2012.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 09/21/2012] [Accepted: 10/10/2012] [Indexed: 11/29/2022]
Abstract
Bacillus anthracis is the causative agent of anthrax and is acquired by three routes of infection: inhalational, gastrointestinal and cutaneous. Gastrointestinal (GI) anthrax is rare, but can rapidly result in severe, systemic disease that is fatal in 25%-60% of cases. Disease mechanisms of GI anthrax remain unclear due to limited numbers of clinical cases and the lack of experimental animal models. Here, we developed an in vivo murine model of GI anthrax where spore survival was maximized through the neutralization of stomach acid followed by an intragastric administration of a thiabendazole paste spore formulation. Infected mice showed a dose-dependent mortality rate and pathological features closely mimicking human GI anthrax. Since Peyer's patches in the murine intestine are the primary sites of B. anthracis growth, we developed a human M (microfold)-like-cell model using a Caco-2/Raji B-cell co-culturing system to study invasive mechanisms of GI anthrax across the intestinal epithelium. Translocation of B. anthracis spores was higher in M-like cells than Caco-2 monolayers, suggesting that M-like cells may serve as an initial entry site for spores. Here, we developed an in vivo murine model of GI anthrax and an in vitro M-like cell model that could be used to further our knowledge of GI anthrax pathogenesis.
Collapse
Affiliation(s)
- Jessica H Tonry
- National Center for Biodefense and Infectious Diseases, George Mason University, 10650 Pyramid Place, Manassas, VA 20110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Immunotherapy, in recent times, has found its application in a variety of immunologically mediated diseases. Oral immunotherapy may not only increase patient compliance but may, in particular, also induce both systemic as well as mucosal immune responses, due to mucosal application of active agents. To improve the bioavailability and to trigger strong immunological responses, recent research projects focused on the encapsulation of drugs and antigens into polymer particles. These particles protect the loaded antigen from the harsh conditions in the GI tract. Furthermore, modification of the surface of particles by the use of lectins, such as Aleuria aurantia lectin, wheatgerm agglutinin or Ulex europaeus-I, enhances the binding to epithelial cells, in particular to membranous cells, of the mucosa-associated lymphoid tissue. Membranous cell-specific targeting leads to an improved transepithelial transport of the particle carriers. Thus, enhanced uptake and presentation of the encapsulated antigen by antigen-presenting cells favor strong systemic, but also local, mucosal immune responses.
Collapse
|
30
|
Hauck CR, Borisova M, Muenzner P. Exploitation of integrin function by pathogenic microbes. Curr Opin Cell Biol 2012; 24:637-44. [PMID: 22884865 DOI: 10.1016/j.ceb.2012.07.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 01/26/2023]
Abstract
Numerous pathogens express adhesive proteins to directly or indirectly associate with integrins. It is well established that by targeting integrins, microbes not only establish an intimate contact with host tissues, but also trigger cellular responses including bacterial internalization. This review will summarize current knowledge about the role of these integrin-dependent processes during infection and how bacteria assure that they efficiently connect to integrins for host cell invasion or translocation of effector molecules. Furthermore, we will discuss recent insight demonstrating that bacteria can harness the physiological, matrix-binding function of integrins for promoting host colonization. From these combined studies, it is becoming evident that integrins are a common nexus for the manipulation of cellular functions by bacterial pathogens. Approaches to disrupt this connection might be an appropriate means to obtain broad-acting tools to modulate a spectrum of infectious diseases.
Collapse
Affiliation(s)
- Christof R Hauck
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Germany.
| | | | | |
Collapse
|
31
|
Keita ÅV, Söderholm JD. Barrier dysfunction and bacterial uptake in the follicle-associated epithelium of ileal Crohn's disease. Ann N Y Acad Sci 2012; 1258:125-34. [DOI: 10.1111/j.1749-6632.2012.06502.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
32
|
Li MF, Hu YH, Zheng WJ, Sun BG, Wang CL, Sun L. Inv1: an Edwardsiella tarda invasin and a protective immunogen that is required for host infection. FISH & SHELLFISH IMMUNOLOGY 2012; 32:586-592. [PMID: 22289712 DOI: 10.1016/j.fsi.2012.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 12/28/2011] [Accepted: 01/13/2012] [Indexed: 05/31/2023]
Abstract
Invasin is an outer membrane protein that is known to mediate entry of enteric bacteria into mammalian cells. In this study, we analyzed the function and immunoprotective potential of the invasin Inv1 from Edwardsiella tarda, a serious fish pathogen that can also infect humans. In silico analysis indicated that Inv1 possesses a conserved N-terminal DUF3442 domain and a C-terminal group 1 bacterial Ig-like domain. Subcellular localization analysis showed that Inv1 is exposed on cell surface and could be recognized by specific antibodies. Mutation of inv1 had no effect on bacterial growth but attenuates overall bacterial virulence and impaired the ability of E. tarda to attach and invade into host cells. Consistent with these observations, antibody blocking of Inv1 inhibited E. tarda infection of host cells. To examine the immunoprotective potential of Inv1, recombinant Inv1 (rInv1) corresponding to the DUF3442 domain was purified and used to vaccinate Japanese flounder (Paralichthys olivaceus). The results showed that rInv1 induced strong protection against lethal-dose challenge of E. tarda. ELISA analysis showed that rInv1-vaccinated fish produced specific serum antibodies that could enhance the serum bactericidal activity against E. tarda. Taken together, these results indicate that Inv1 is a surface-localized virulence factor that is involved in host infection and can induce effective immunoprotection when used as a subunit vaccine.
Collapse
Affiliation(s)
- Mo-fei Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao 266071, China
| | | | | | | | | | | |
Collapse
|
33
|
Mahler GJ, Esch MB, Tako E, Southard TL, Archer SD, Glahn RP, Shuler ML. Oral exposure to polystyrene nanoparticles affects iron absorption. NATURE NANOTECHNOLOGY 2012; 7:264-71. [PMID: 22327877 DOI: 10.1038/nnano.2012.3] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/06/2012] [Indexed: 05/18/2023]
Abstract
The use of engineered nanoparticles in food and pharmaceuticals is expected to increase, but the impact of chronic oral exposure to nanoparticles on human health remains unknown. Here, we show that chronic and acute oral exposure to polystyrene nanoparticles can influence iron uptake and iron transport in an in vitro model of the intestinal epithelium and an in vivo chicken intestinal loop model. Intestinal cells that are exposed to high doses of nanoparticles showed increased iron transport due to nanoparticle disruption of the cell membrane. Chickens acutely exposed to carboxylated particles (50 nm in diameter) had a lower iron absorption than unexposed or chronically exposed birds. Chronic exposure caused remodelling of the intestinal villi, which increased the surface area available for iron absorption. The agreement between the in vitro and in vivo results suggests that our in vitro intestinal epithelium model is potentially useful for toxicology studies.
Collapse
Affiliation(s)
- Gretchen J Mahler
- Department of Bioengineering, Binghamton University, Binghamton, New York 13902, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Giuliano EA, Finn K. Characterization of membranous (M) cells in normal feline conjunctiva-associated lymphoid tissue (CALT). Vet Ophthalmol 2012; 14 Suppl 1:60-6. [PMID: 21923825 DOI: 10.1111/j.1463-5224.2011.00917.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To characterize conjunctival lymphoid nodules obtained from the nictitans of healthy cats to determine if the follicle-associated epithelium (FAE) of conjunctiva-associated lymphoid tissue (CALT) in this species contains membranous (M)-cells analogous to those described in other regions of mucosa-associated lymphoid tissue (MALT). METHODS Lymphoid follicles from nictitan bulbar surfaces of 10 healthy cats (20 eyes total) were examined. Nictitans from five cats were harvested immediately post-mortem and a minimum of 12 lymphoid nodules from each third eyelid were isolated using a Zeiss operating microscope. At least three lymphoid follicles from each eye were examined using light microscopy (LM), transmission electron microscopy (TEM), and scanning electron microscopy (SEM) using standard fixation and embedding protocols. Nictitan-lymphoid follicles from another five healthy cats were processed for immunohistochemistry to characterize the distribution of T- and B-lymphocytes present beneath the FAE. RESULTS The FAE overlying CALT from 10 healthy cats demonstrated morphology characteristic of M-cells including attenuated apical cell surface with blunted microvilli and microfolds, invaginated basolateral membrane forming a cytoplasmic pocket, and diminished distance between the apical and pocket membrane. Immunohistochemistry of lymphoid tissue subtending the FAE demonstrated B-cell dependent regions in the germinal centers surrounded by T-cell dependent interfollicular zones. CONCLUSIONS Healthy feline CALT contains morphologic features analogous to those described in other regions of MALT. Documentation of feline conjunctival M-cells is of clinical relevance in the study of primary infectious, allergic, and autoimmune ocular diseases, as well as a potential means of vaccination or drug delivery.
Collapse
Affiliation(s)
- Elizabeth A Giuliano
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA.
| | | |
Collapse
|
35
|
Fàbrega A, Vila J. Yersinia enterocolitica: Pathogenesis, virulence and antimicrobial resistance. Enferm Infecc Microbiol Clin 2012; 30:24-32. [DOI: 10.1016/j.eimc.2011.07.017] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 07/12/2011] [Accepted: 07/15/2011] [Indexed: 12/29/2022]
|
36
|
Kim SH, Jung DI, Yang IY, Kim J, Lee KY, Nochi T, Kiyono H, Jang YS. M cells expressing the complement C5a receptor are efficient targets for mucosal vaccine delivery. Eur J Immunol 2011; 41:3219-29. [PMID: 21887786 DOI: 10.1002/eji.201141592] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 07/21/2011] [Accepted: 08/26/2011] [Indexed: 11/10/2022]
Abstract
In the mucosal immune system, M cells are known as specialized epithelial cells that take up luminal antigens, although the receptors on M cells and the mechanism of antigen uptake into M cells are not well-understood. Here, we report the expression of the complement C5a receptor (C5aR) on the apical surface of M cells. C5ar mRNA expression in co-cultured Caco-2 human M-like cells was six-fold higher than in mono-cultured cells. C5aR expression was detected together with glycoprotein 2, an M-cell-specific protein, on the apical surface of M-like cells and mouse Peyer's patch M cells. Interestingly, after oral administration of Yersinia enterocolitica which expresses outer membrane protein H (OmpH) that is homologous to the Skp α1 domain of Escherichia coli, a ligand of C5aR, dense clustering and phosphorylation of C5aR were detected in M cells. Finally, targeted antigen delivery to M cells using C5aR as a receptor was achieved using the OmpH α1 of Y. enterocolitica such that the induction of ligand-conjugated antigen-specific immune responses was confirmed in mice after oral immunization of the OmpH β1α1-conjugated antigen. Collectively, we identified C5aR expression on M cells and suggest that C5aR could be used as a target receptor for mucosal antigen delivery.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology and Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Trcek J, Berschl K, Trülzsch K. In vivo analysis of Yersinia enterocolitica infection using luxCDABE. FEMS Microbiol Lett 2010; 307:201-6. [PMID: 20636977 DOI: 10.1111/j.1574-6968.2010.01983.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Yersiniae expressing an L-arabinose-inducible luxCDABE reporter were used to analyze the colonization of mice. Infection of live mice was followed over a period of 6 days. These experiments revealed frequent colonization of cervical lymph nodes after oral, but not intravenous infection. Furthermore, the well-known colonization of the small intestine, Peyer's patches (PPs) of the ileum, the cecal lymph follicle, mesenteric lymph nodes, liver, and spleen was easily detectable. Removal of the intestinal tract of mice revealed that the number of abscessed PPs and other tissues can be easily quantified. Experiments with an invasin mutant expressing luxCDABE revealed a significantly reduced number of abscessed PPs, cecal lymph follicles, and lymph nodes in yersiniae lacking invasin.
Collapse
Affiliation(s)
- Janja Trcek
- Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Ludwig Maximilans Universität, Munich, Germany
| | | | | |
Collapse
|
38
|
Friswell M, Campbell B, Rhodes J. The role of bacteria in the pathogenesis of inflammatory bowel disease. Gut Liver 2010; 4:295-306. [PMID: 20981205 DOI: 10.5009/gnl.2010.4.3.295] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 04/06/2010] [Indexed: 12/19/2022] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) have features that suggest bacterial involvement, and all genetic models of inflammatory bowel disease (IBD) require the presence of commensal bacteria. CD is associated with innate immune response genes such as NOD2/CARD15 and the autophagy genes ATG16L1 and IRGM. However, IBD responds to immunosuppression, suggesting that any bacteria involved are not acting as conventional pathogens. Molecular techniques are rapidly advancing our knowledge of the gut microbiota. In CD there is reduced diversity, and notably a reduction in the probiotic Faecalibacterium prausnitzii, the presence of which in the terminal ileum is associated with a reduced risk of recurrence following surgery. There is also a consistent increase in mucosa-associated Escherichia coli with an "adherent and invasive" phenotype, which allows them to replicate inside macrophages and induce granulomas. Speculation that CD could be caused by the Mycobacterium avium subspecies paratuberculosis (MAP) continues. The response to antitumor necrosis factor treatments suggests that, if relevant at all, MAP is not acting as a conventional pathogen. However, there is increased colonization by MAP in CD, and there is evidence that it could have an indirect effect mediated by the suppression of macrophage function. UC relapse is frequently associated with infection by pathogens, but there is less evidence for involvement of a specific bacterial species. Poor barrier integrity followed by an inflammatory reaction to bacterial components, with chronicity maintained by an autoimmune process, seems a plausible pathogenic model. Bacterial theories of pathogenesis are now becoming testable by targeted therapeutic interventions.
Collapse
Affiliation(s)
- Melissa Friswell
- Gastroenterology Research Unit, University of Liverpool School of Clinical Sciences, Liverpool, UK
| | | | | |
Collapse
|
39
|
Edelmann MJ, Kramer HB, Altun M, Kessler BM. Post-translational modification of the deubiquitinating enzyme otubain 1 modulates active RhoA levels and susceptibility to Yersinia invasion. FEBS J 2010; 277:2515-30. [PMID: 20553488 DOI: 10.1111/j.1742-4658.2010.07665.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microbial pathogens exploit the ubiquitin system to facilitate infection and manipulate the immune responses of the host. In this study, susceptibility to Yersinia enterocolitica and Yersinia pseudotuberculosis invasion was found to be increased upon overexpression of the deubiquitinating enzyme otubain 1 (OTUB1), a member of the ovarian tumour domain-containing protein family. Conversely, OTUB1 knockdown interfered with Yersinia invasion in HEK293T cells as well as in primary monocytes. This effect was attributed to a modulation of bacterial uptake. We demonstrate that the Yersinia-encoded virulence factor YpkA (YopO) kinase interacts with a post-translationally modified form of OTUB1 that contains multiple phosphorylation sites. OTUB1, YpkA and the small GTPase ras homologue gene family member A (RhoA) were found to be part of the same protein complex, suggesting that RhoA levels are modulated by OTUB1. Our results show that OTUB1 is able to stabilize active RhoA prior to invasion, which is concomitant with an increase in bacterial uptake. This effect is modulated by post-translational modifications of OTUB1, suggesting a new entry point for manipulating Yersinia interactions with the host.
Collapse
|
40
|
Keita AV, Söderholm JD, Ericson AC. Stress-induced barrier disruption of rat follicle-associated epithelium involves corticotropin-releasing hormone, acetylcholine, substance P, and mast cells. Neurogastroenterol Motil 2010; 22:770-8, e221-2. [PMID: 20149111 DOI: 10.1111/j.1365-2982.2010.01471.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The follicle-associated epithelium (FAE) is specialized in uptake and sampling of luminal antigens and bacteria. We previously showed that stress increased FAE permeability in rats. An increased uptake may alter antigen exposure in Peyer's patches leading to intestinal disease. The aim of this study was to elucidate mechanisms involved in the acute stress-induced increase in FAE permeability. METHODS Rats were pretreated i.p. with corticotropin-releasing hormone receptor (CRH-R) antagonist, neurokinin receptor 1 (NK-1R) antagonist, atropine, the mast cell stabilizer doxantrazole (DOX), or NaCl, and submitted to 1-h acute water avoidance stress. FAE tissues were mounted in Ussing chambers for measurements of permeability to (51)Cr-EDTA, horseradish peroxidase (HRP) and chemically killed Escherichia coli K-12. Further, FAE segments were exposed in vitro in chambers to CRH, substance P (SP), carbachol, and DOX. Neurotransmitter- and receptor distribution was studied by immunohistochemistry. KEY RESULTS Stress-induced increases in uptake across FAE of HRP and E. coli were reduced by DOX, CRH-R antagonist and atropine, whereas the NK-1R antagonist decreased (51)Cr-EDTA permeability. Exposure to CRH and carbachol increased HRP and E. coli passage, whereas SP increased bacterial and (51)Cr-EDTA permeability. DOX counteracted all of these effects. Immunohistochemistry revealed CRH, acetylcholine, SP, and their receptors on mast cells within the Peyer's patches, subepithelial dome, and adjacent villi. CONCLUSIONS & INFERENCES Corticotropin-releasing hormone and acetylcholine signaling affect mainly transcellular permeability while SP seems more selective toward the paracellular pathways. Our findings may be of importance for the understanding of the pathogenesis of stress-related intestinal disorders.
Collapse
Affiliation(s)
- A V Keita
- Division of Surgery and Clinical Oncology, Department of Clinical and Experimental Medicine, Clinical and Experimental Research, Faculty of Health Science, University Hospital, Linköping, Sweden
| | | | | |
Collapse
|
41
|
Uliczka F, Kornprobst T, Eitel J, Schneider D, Dersch P. Cell invasion of Yersinia pseudotuberculosis by invasin and YadA requires protein kinase C, phospholipase C-gamma1 and Akt kinase. Cell Microbiol 2009; 11:1782-801. [PMID: 19681907 DOI: 10.1111/j.1462-5822.2009.01371.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The outer membrane proteins YadA and invasin of Yersinia pseudotuberculosis promote invasion into mammalian cells through beta(1)-integrins and trigger the production of interleukin (IL)-8. FAK, c-Src and the PI3 kinase were previously found to be important for both YadA- and invasin-promoted uptake. Here, we demonstrate that two different downstream effectors of PI3 kinase, Akt and phospholipase Cgamma1 are required for efficient cell invasion. Inhibition of Akt or phospholipase C-gamma (PLC-gamma)1 by pharmaceutical agents as well as reduced expression of the isoforms Akt1 and Akt2, and of PLC-gamma1 by RNA interference decreased entry of YadA- and Inv-expressing bacteria significantly. In addition, we report that the conventional protein kinases C (PKC)alpha and -beta, positioned downstream of PLC-gamma1, are activated upon Inv- or YadA-promoted cell entry. They colocalize with intracellular bacteria and their depletion by siRNA treatment also resulted in a strong reduction of cell entry. In contrast, neither Akt nor PLC-gamma1, and the PKCs are essential for YadA- and Inv-mediated IL-8 synthesis and release. We conclude that YadA and invasin of Y. pseudotuberculosis both trigger similar signal transduction pathways during integrin-mediated phagocytosis into epithelial cells, which lead to the activation of Akt, PLC-gamma1, PKCalpha and -beta downstream of PI3 kinase, separate from the MAPK-dependent pathway that triggers IL-8 production.
Collapse
Affiliation(s)
- Frank Uliczka
- Institut für Mikrobiologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | | | | | | | | |
Collapse
|
42
|
Bacterial particle endocytosis by epithelial cells is selective and enhanced by tumor necrosis factor receptor ligands. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:397-407. [PMID: 19129472 DOI: 10.1128/cvi.00210-08] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Bacterial pathogens use virulence strategies to invade epithelial barriers, but active processes of epithelial cells may also contribute to the endocytosis of microbial particles. To focus on the latter, we studied the uptake of fixed and fluorescently labeled bacterial particles in intestinal and bronchoepithelial cell cultures and found it to be enhanced in Caco-2BBe and NCI-H292 cells after treatment with tumor necrosis factor alpha and an agonist antibody against the lymphotoxin beta receptor. Confocal fluorescence microscopy, flow cytometry, and transmission electron microscopy revealed that Staphylococcus aureus and Yersinia enterocolitica were readily endocytosed, although there was scant uptake of Shigella sonnei, Salmonella enterica serovar Typhimurium, and Klebsiella pneumoniae particles. Endocytosed Staphylococcus was often associated with cytoplasmic claudin-4 vesicles; this was not found for Yersinia, suggesting that cytokine treatment upregulated two distinct endocytosis pathways. Interestingly, when Staphylococcus and Yersinia were coincubated with epithelial monolayers, the cells were unlikely to take up Yersinia unless they had also endocytosed large numbers of Staphylococcus particles, although the two bacteria were apparently processed in distinct compartments. Cytokine treatment induced an upregulation and redistribution of beta1 integrin to the apical surface of NCI-H292 cells; consistent with this effect, treatment with anti-beta1 integrin antibody blocked uptake of both Yersinia and Staphylococcus in NCI-H292 and Caco-2BBe cells. Our results suggest that capture of bacterial particles by mucosal epithelial cells is selective and that different endocytic mechanisms are enhanced by proinflammatory cytokines.
Collapse
|
43
|
Yersinia pseudotuberculosis induces transcytosis of nanoparticles across human intestinal villus epithelium via invasin-dependent macropinocytosis. J Transl Med 2008; 88:1215-26. [PMID: 18810251 DOI: 10.1038/labinvest.2008.86] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Crohn's disease is characterized by a defect in intestinal barrier function, where bacteria are considered the most important inflammation-driving factor. Enteric bacteria, including E. coli and Yersinia spp, affect tight junctions in enterocytes, but little is known about bacterial effects on the transcellular pathway. Our objective was to study the short-term effects of Y. pseudotuberculosis on uptake of nanoparticles across human villus epithelium. Monolayers of human colon epithelium-derived Caco-2 cells and biopsies of normal human ileum were studied after 2 h exposure to Y. pseudotuberculosis expressing (inv+) or lacking (inv-) the bacterial adhesion molecule, invasin. Transepithelial transport of fluorescent nanoparticles (markers of transcytosis) was quantified by flow cytometry, and mechanisms explored by using inhibitors of endocytosis. Epithelial expressions of beta1-integrin and particle uptake pathways were studied by confocal microscopy. The paracellular pathway was assessed by electrical resistance (TER), mannitol flux, and expression of tight junction proteins occludin and caludin-4 by confocal microscopy. Inv+ Y. pseudotuberculosis adhered to the apical surface of epithelial cells and induced transcytosis of exogenous nanoparticles across Caco-2 monolayers (30-fold increase, P<0.01) and ileal mucosa (268+/-47% of control; P<0.01), whereas inv bacteria had no effect on transcytosis. The transcytosis was concentration-, particle size- and temperature-dependent, and possibly mediated via macropinocytosis. Y. pseudotuberculosis also induced apical expression of beta1-integrin on epithelial cells. A slight drop in TER was seen after exposure to inv+ Y. pseudotuberculosis, whereas mannitol flux and tight junction protein expression was unchanged. In summary, Y. pseudotuberculosis induced apical expression of beta1-integrin and stimulated uptake of nanoparticles via invasin-dependent transcytosis in human intestinal epithelium. Our findings suggest that bacterial factors may initiate transcytosis of luminal exogenous particles across human ileal mucosa, thus presenting a novel mechanism of intestinal barrier dysfunction.
Collapse
|
44
|
Abstract
BACKGROUND M (microfold or membranous) cells are specialised epithelial cells responsible for antigen sampling at the interface of mucosal surfaces and the environment. Their high transcytotic ability make M cells an attractive target for mucosally delivered vaccines and therapeutics. OBJECTIVE This brief review discusses the current state of M cell-targeted mucosal delivery systems and the potential of such delivery systems for the development of new vaccines and therapeutics against mucosal infectious and inflammatory diseases. SCOPE A variety of synthetic microparticles/nanoparticles have been developed and tested as vehicles for M cell-targeted mucosal drug and vaccine delivery. beta1 integrins, pathogen recognition receptors, specific carbohydrate residues and other M cell surface antigens have been exploited as potential targets for the delivery of mucosal vaccines and therapeutics. CONCLUSION Despite a considerable body of literature, much work still needs to be done before an effective M cell-targeted vaccine or therapeutic is developed.
Collapse
Affiliation(s)
- Rhonda Kuolee
- Institute for Biological Sciences, National Research Council Canada, Ottawa, Ontario K1A 0R6, Canada
| | | |
Collapse
|
45
|
Mantis NJ, Wagner J. Analysis of Adhesion Molecules Involved in Leukocyte Homing into the Basolateral Pockets of Mouse Peyer's Patch M Cells. J Drug Target 2008; 12:79-87. [PMID: 15203901 DOI: 10.1080/10611860410001693724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The basolateral membranes of intestinal M cells are invaginated to form large intraepithelial "pockets" that are populated by specific sub-sets of mucosal leukocytes, including CD4+ T cells, memory and naïve B cells, and occasional dendritic cells. The adhesion molecules involved in leukocyte trafficking and/or retention within this unique immunological niche are unknown. In this study, we used immunofluorescence microscopy and a battery of monoclonal antibodies to identify the adhesion molecules expressed by leukocytes situated within the intracellular pockets of mouse Peyer's patch (PP) M cells. M cell associated leukocytes (MAL) consistently stained positive for integrin alpha4beta7, and integrin LFA-1 (CD11a/CD18), but were rarely positive for L-selectin (CD62L) or the mucosal integrin alphaEbeta7. However, neither the alpha4beta7 ligands MadCAM-1 or VCAM-1, nor the LFA-1 ligand ICAM-1, were detected on M cell basolateral membranes. To determine whether integrins alpha4beta7 or LFA-1 play a functional role leukocyte homing to M cell pockets, we examined M cells in mice deficient in integrin beta7 or CD11a/CD18. Although PP from CD18 or integrin beta7 mice were reduced in number and size as compared to age-matched controls, we identified M cells in both strains of mice. However, mice lacking CD18 (but not integrin beta7) had significantly fewer leukocytes within M cell pockets as compared to control animals, suggesting LFA-1 (but not alpha4beta7) may contribute, in part, to leukocyte trafficking into and/or retention within this unique immunological niche.
Collapse
Affiliation(s)
- Nicholas J Mantis
- GI Cell Biology Laboratory, Children's Hospital Boston, 300 Longwood Avenue, 02115 Boston, MA, USA.
| | | |
Collapse
|
46
|
Corr SC, Gahan CCGM, Hill C. M-cells: origin, morphology and role in mucosal immunity and microbial pathogenesis. ACTA ACUST UNITED AC 2007; 52:2-12. [PMID: 18081850 DOI: 10.1111/j.1574-695x.2007.00359.x] [Citation(s) in RCA: 189] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
M-cells are specialized cells found in the follicle-associated epithelium of intestinal Peyer's patches of gut-associated lymphoid tissue and in isolated lymphoid follicles, appendix and in mucosal-associated lymphoid tissue sites outside the gastrointestinal tract. In the gastrointestinal tract, M-cells play an important role in transport of antigen from the lumen of the small intestine to mucosal lymphoid tissues, where processing and initiation of immune responses occur. Thus, M-cells act as gateways to the mucosal immune system and this function has been exploited by many invading pathogens. Understanding the mechanism by which M-cells sample antigen will inform the design of oral vaccines with improved efficacy in priming mucosal and systemic immune responses. In this review, the origin and morphology of M-cells, and their role in mucosal immunity and pathogenesis of infections are discussed.
Collapse
Affiliation(s)
- Sinead C Corr
- Department of Microbiology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.
| | | | | |
Collapse
|
47
|
A conserved glycine residue of trimeric autotransporter domains plays a key role in Yersinia adhesin A autotransport. J Bacteriol 2007; 189:9011-9. [PMID: 17921300 DOI: 10.1128/jb.00985-07] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The Yersinia adhesin A (YadA) is a trimeric autotransporter adhesin of enteric yersiniae. It consists of three major domains: a head mediating adherence to host cells, a stalk involved in serum resistance, and an anchor that forms a membrane pore and is responsible for the autotransport function. The anchor contains a glycine residue, nearly invariant throughout trimeric autotransporter adhesins, that faces the pore lumen. To address the role of this glycine, we replaced it with polar amino acids of increasing side chain size and expressed wild-type and mutant YadA in Escherichia coli. The mutations did not impair the YadA-mediated adhesion to collagen and to host cells or the host cell cytokine production, but they decreased the expression levels and stability of YadA trimers with increasing side chain size. Likewise, autoagglutination and resistance to serum were decreased in these mutants. We found that the periplasmic protease DegP is involved in the degradation of YadA and that in an E. coli degP deletion strain, mutant versions of YadA were expressed almost to wild-type levels. We conclude that the conserved glycine residue affects both the export and the stability of YadA and consequently some of its putative functions in pathogenesis.
Collapse
|
48
|
Autenrieth SE, Autenrieth IB. Yersinia enterocolitica: subversion of adaptive immunity and implications for vaccine development. Int J Med Microbiol 2007; 298:69-77. [PMID: 17702651 DOI: 10.1016/j.ijmm.2007.07.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Enteric Yersinia spp. invade Peyer's patches, disseminate to lymphoid tissues, and induce mucosal and systemic immune responses. Many virulence factors of Yersinia enterocolitica have been investigated in detail and were found to act on host cells involved in innate and adaptive immunity. Recent work explored as to whether attenuated Y. enterocolitica or recombinant components of Y. enterocolitica can be used as tools for vaccination. We and others have tested whether by means of the type three secretion system in attenuated Y. enterocolitica strains antigens might be delivered to antigen-presenting cells in order to induce CD8 and CD4 T cell responses. Alternatively, recombinant components of Y. enterocolitica such as invasin protein which binds to beta1 integrins of host cells have been tested for their ability to target antigen along with microparticles (fused to invasin) to antigen-presenting cells and to act as adjuvant. The work summarized in this article demonstrates that Y. enterocolitica and its components might be useful tools for novel vaccination strategies; in fact, invasin when fused to antigen and coated to microparticles might induce both CD4 and CD8 T cell responses. Likewise, attenuated Y. enterocolitica live carrier strains were reported to induce both CD8 and some CD4 T cell responses. However, we need to know more about how Y. enterocolitica subverts functions of antigen-presenting cells in order to design mutants with optimized antigen delivery features and deletion in those virulence factor that contribute to subversion of innate or adaptive immune responses.
Collapse
Affiliation(s)
- Stella E Autenrieth
- Institut für Medizinische Mikrobiologie und Hygiene, Universitätsklinikum Tübingen, Elfriede-Aulhorn-Strasse 6, D-72076 Tübingen, Germany
| | | |
Collapse
|
49
|
Lawrenz MB, Miller VL. Comparative analysis of the regulation of rovA from the pathogenic yersiniae. J Bacteriol 2007; 189:5963-75. [PMID: 17573476 PMCID: PMC1952055 DOI: 10.1128/jb.00528-07] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
RovA is a MarR/SlyA-type regulator that mediates the transcription of inv in Yersinia enterocolitica and Y. pseudotuberculosis. In Y. pseudotuberculosis, rovA transcription is controlled primarily by H-NS and RovA, which bind to similar regions within the rovA promoter. At 37 degrees C, rovA transcription is repressed by H-NS. Transcription of rovA results when RovA relieves H-NS-mediated repression. The region of the rovA promoter that H-NS and RovA bind is not conserved in the Y. enterocolitica promoter. Using green fluorescent protein reporters, we determined that the Y. enterocolitica rovA (rovA(Yent)) promoter is weaker than the Y. pseudotuberculosis promoter. However, despite the missing H-NS/RovA binding site in the rovA(Yent) promoter, H-NS and RovA are still involved in the regulation of rovA(Yent). DNA binding studies suggest that H-NS and RovA bind with a higher affinity to the Y. pseudotuberculosis/Y. pestis rovA (rovA(Ypstb/Ypestis)) promoter than to the rovA(Yent) promoter. Furthermore, H-NS appears to bind to two regions in a cooperative fashion within the rovA(Yent) promoter that is not observed with the rovA(Ypstb/Ypestis) promoter. Finally, using a transposon mutagenesis approach, we identified a new positive regulator of rovA in Y. enterocolitica, LeuO. In Escherichia coli, LeuO regulates gene expression via changes in levels of RpoS and H-NS, but LeuO-mediated regulation of rovA(Yent) appears to be independent of either of these two proteins. Together, these data demonstrate that while the rovA regulatory factors are conserved in Yersinia, divergence of Y. enterocolitica and Y. pseudotuberculosis/Y. pestis during evolution has resulted in modifications in the mechanisms that are responsible for controlling rovA transcription.
Collapse
Affiliation(s)
- Matthew B Lawrenz
- Washington University School of Medicine, Department of Molecular Microbiology, 660 S. Euclid Avenue, Campus Box 8230, St. Louis, MO 63110, USA.
| | | |
Collapse
|
50
|
Martinez-Argudo I, Sands C, Jepson MA. Translocation of enteropathogenic Escherichia coli across an in vitro M cell model is regulated by its type III secretion system. Cell Microbiol 2007; 9:1538-46. [PMID: 17298392 DOI: 10.1111/j.1462-5822.2007.00891.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC) is an extracellular pathogen that utilizes a type III secretion system (TTSS) to modulate diverse host cell processes including cytoskeletal dynamics, tight junction permeability and macrophage phagocytosis. Some EPEC strains exhibit selective tropism for the specialized follicle-associated epithelium (FAE) overlying lymphoid follicles in the gut, which is a major site of uptake of inert particulates and pathogens, but do not translocate from the intestinal lumen in significant numbers. We have investigated the interaction of EPEC with FAE using an established in vitro model of the specialized FAE in which polarized enterocyte-like Caco-2 cells cocultured with the Raji B cell line undergo a phenotypic switch to a form that morphologically and functionally resembles the specialized antigen-transporting M cells found within FAE. Having confirmed that coculture with Raji B cells induces brush border reorganization and enhances particle transport across Caco-2 cells, we investigated translocation of bacteria across the M cell model. While Salmonella translocation was markedly upregulated by Raji coculture, transport of wild-type EPEC occurred at similarly low levels across both native Caco-2 and Caco-2/Raji-cocultured layers. Translocation rates were markedly higher for EPEC strains lacking either functional TTSS or the effector protein EspF. These observations resemble previously reported data on the inhibition of macrophage phagocytosis by EPEC, which has also been reported to be dependent on TTSS and EspF. Furthermore, as with macrophage phagocytosis, enhanced translocation of a TTSS mutant was blocked by wortmannin, implicating inhibition of phosphatidyl inositol 3-kinase-mediated signalling in the regulation of M cell translocation by EPEC.
Collapse
Affiliation(s)
- Isabel Martinez-Argudo
- Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | | | | |
Collapse
|