1
|
Campylobacter jejuni Triggers Signaling through Host Cell Focal Adhesions To Inhibit Cell Motility. mBio 2021; 12:e0149421. [PMID: 34425711 PMCID: PMC8406305 DOI: 10.1128/mbio.01494-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Campylobacter jejuni is a major foodborne pathogen that exploits the focal adhesions of intestinal cells to promote invasion and cause severe gastritis. Focal adhesions are multiprotein complexes involved in bidirectional signaling between the actin cytoskeleton and the extracellular matrix. We investigated the dynamics of focal adhesion structure and function in C. jejuni-infected cells using a comprehensive set of approaches, including confocal microscopy of live and fixed cells, immunoblotting, and superresolution interferometric photoactivated localization microscopy (iPALM). We found that C. jejuni infection of epithelial cells results in increased focal adhesion size and altered topology. These changes resulted in a persistent modulatory effect on the host cell focal adhesion, evidenced by an increase in cell adhesion strength, a decrease in individual cell motility, and a reduction in collective cell migration. We discovered that C. jejuni infection causes an increase in phosphorylation of paxillin and an alteration of paxillin turnover at the focal adhesion, which together represent a potential mechanistic basis for altered cell motility. Finally, we observed that infection of epithelial cells with the C. jejuni wild-type strain in the presence of a protein synthesis inhibitor, a C. jejuni CadF and FlpA fibronectin-binding protein mutant, or a C. jejuni flagellar export mutant blunts paxillin phosphorylation and partially reestablishes individual host cell motility and collective cell migration. These findings provide a potential mechanism for the restricted intestinal repair observed in C. jejuni-infected animals and raise the possibility that bacteria targeting extracellular matrix components can alter cell behavior after binding and internalization by manipulating focal adhesions.
Collapse
|
2
|
Sipos A, Ujlaki G, Mikó E, Maka E, Szabó J, Uray K, Krasznai Z, Bai P. The role of the microbiome in ovarian cancer: mechanistic insights into oncobiosis and to bacterial metabolite signaling. Mol Med 2021; 27:33. [PMID: 33794773 PMCID: PMC8017782 DOI: 10.1186/s10020-021-00295-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is characterized by dysbiosis, referred to as oncobiosis in neoplastic diseases. In ovarian cancer, oncobiosis was identified in numerous compartments, including the tumor tissue itself, the upper and lower female genital tract, serum, peritoneum, and the intestines. Colonization was linked to Gram-negative bacteria with high inflammatory potential. Local inflammation probably participates in the initiation and continuation of carcinogenesis. Furthermore, local bacterial colonies in the peritoneum may facilitate metastasis formation in ovarian cancer. Vaginal infections (e.g. Neisseria gonorrhoeae or Chlamydia trachomatis) increase the risk of developing ovarian cancer. Bacterial metabolites, produced by the healthy eubiome or the oncobiome, may exert autocrine, paracrine, and hormone-like effects, as was evidenced in breast cancer or pancreas adenocarcinoma. We discuss the possible involvement of lipopolysaccharides, lysophosphatides and tryptophan metabolites, as well as, short-chain fatty acids, secondary bile acids and polyamines in the carcinogenesis of ovarian cancer. We discuss the applicability of nutrients, antibiotics, and probiotics to harness the microbiome and support ovarian cancer therapy. The oncobiome and the most likely bacterial metabolites play vital roles in mediating the effectiveness of chemotherapy. Finally, we discuss the potential of oncobiotic changes as biomarkers for the diagnosis of ovarian cancer and microbial metabolites as possible adjuvant agents in therapy.
Collapse
Affiliation(s)
- Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Gyula Ujlaki
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Eszter Maka
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, Debrecen, 4032, Hungary
| | - Judit Szabó
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Zoárd Krasznai
- Department of Gynecology and Obstetrics, Faculty of Medicine, University of Debrecen, Egyetem tér 1, Debrecen, 4032, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, 4032, Hungary.
- Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary.
| |
Collapse
|
3
|
Abstract
Campylobacter jejuni and Campylobacter coli can be frequently isolated from poultry and poultry-derived products, and in combination these two species cause a large portion of human bacterial gastroenteritis cases. While birds are typically colonized by these Campylobacter species without clinical symptoms, in humans they cause (foodborne) infections at high frequencies, estimated to cost billions of dollars worldwide every year. The clinical outcome of Campylobacter infections comprises malaise, diarrhea, abdominal pain and fever. Symptoms may continue for up to two weeks and are generally self-limiting, though occasionally the disease can be more severe or result in post-infection sequelae. The virulence properties of these pathogens have been best-characterized for C. jejuni, and their actions are reviewed here. Various virulence-associated bacterial determinants include the flagellum, numerous flagellar secreted factors, protein adhesins, cytolethal distending toxin (CDT), lipooligosaccharide (LOS), serine protease HtrA and others. These factors are involved in several pathogenicity-linked properties that can be divided into bacterial chemotaxis, motility, attachment, invasion, survival, cellular transmigration and spread to deeper tissue. All of these steps require intimate interactions between bacteria and host cells (including immune cells), enabled by the collection of bacterial and host factors that have already been identified. The assortment of pathogenicity-associated factors now recognized for C. jejuni, their function and the proposed host cell factors that are involved in crucial steps leading to disease are discussed in detail.
Collapse
|
4
|
Šikić Pogačar M, Langerholc T, Mičetić-Turk D, Možina SS, Klančnik A. Effect of Lactobacillus spp. on adhesion, invasion, and translocation of Campylobacter jejuni in chicken and pig small-intestinal epithelial cell lines. BMC Vet Res 2020; 16:34. [PMID: 32013961 PMCID: PMC6998324 DOI: 10.1186/s12917-020-2238-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/09/2020] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Campylobacter spp. are a major cause of bacterial food-borne diarrhoeal disease. This mainly arises through contamination of meat products during processing. For infection, Campylobacter spp. must adhere to epithelial cells of the mucus layer, survive conditions of the gastrointestinal tract, and colonise the intestine of the host. Addition of probiotic bacteria might promote competitive adhesion to epithelial cells, consequently reducing Campylobacter jejuni colonisation. Effect of Lactobacillus spp. (PCS20, PCS22, PCS25, LGG, PCK9) on C. jejuni adhesion, invasion and translocation in pig (PSI cl.1) and chicken (B1OXI) small-intestine cell lines, as well as pig enterocytes (CLAB) was investigated. RESULTS Overall, in competitive adhesion assays with PSI cl.1 and CLAB cell monolayers, the addition of Lactobacillus spp. reduced C. jejuni adherence to the cell surface, and negatively affected the C. jejuni invasion. Interestingly, Lactobacillus spp. significantly impaired C. jejuni adhesion in three-dimensional functional PSI cl.1 and B1OXI cell models. Also, C. jejuni did not translocate across PSI cl.1 and B1OXI cell monolayers when co-incubated with probiotics. Among selected probiotics, Lactobacillus rhamnosus LGG was the strain that reduced adhesion efficacy of C. jejuni most significantly under co-culture conditions. CONCLUSION The addition of Lactobacillus spp. to feed additives in livestock nutrition might be an effective novel strategy that targets Campylobacter adhesion to epithelial cells, and thus prevents colonisation, reduces the transmission, and finally lowers the incidence of human campylobacteriosis.
Collapse
Affiliation(s)
| | - Tomaž Langerholc
- Department of Microbiology, Biochemistry, Molecular Biology and Biotechnology, Faculty of Agriculture and Life Science, University of Maribor, 2311, Hoče, Slovenia
| | | | - Sonja Smole Možina
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Anja Klančnik
- Department of Food Science and Technology, Biotechnical Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| |
Collapse
|
5
|
Federman C, Ma C, Biswas D. Major components of orange oil inhibit Staphylococcus aureus growth and biofilm formation, and alter its virulence factors. J Med Microbiol 2016; 65:688-695. [DOI: 10.1099/jmm.0.000286] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Cassandra Federman
- Department of Animal and Avian Sciences, University of Maryland, College Park, USA
| | - Christopher Ma
- Department of Animal and Avian Sciences, University of Maryland, College Park, USA
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, USA
- Center for Food Safety and Security Systems, University of Maryland, College Park MD 20742, USA
| |
Collapse
|
6
|
Saint-Cyr MJ, Guyard-Nicodème M, Messaoudi S, Chemaly M, Cappelier JM, Dousset X, Haddad N. Recent Advances in Screening of Anti-Campylobacter Activity in Probiotics for Use in Poultry. Front Microbiol 2016; 7:553. [PMID: 27303366 PMCID: PMC4885830 DOI: 10.3389/fmicb.2016.00553] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/04/2016] [Indexed: 12/17/2022] Open
Abstract
Campylobacteriosis is the most common cause of bacterial gastroenteritis worldwide. Campylobacter species involved in this infection usually include the thermotolerant species Campylobacter jejuni. The major reservoir for C. jejuni leading to human infections is commercial broiler chickens. Poultry flocks are frequently colonized by C. jejuni without any apparent symptoms. Risk assessment analyses have identified the handling and consumption of poultry meat as one of the most important sources of human campylobacteriosis, so elimination of Campylobacter in the poultry reservoir is a crucial step in the control of this foodborne infection. To date, the use of probiotics has demonstrated promising results to reduce Campylobacter colonization. This review provides recent insights into methods used for probiotic screening to reduce the prevalence and colonization of Campylobacter at the farm level. Different eukaryotic epithelial cell lines are employed to screen probiotics with an anti-Campylobacter activity and yield useful information about the inhibition mechanism involved. These in vitro virulence models involve only human intestinal or cervical cell lines whereas the use of avian cell lines could be a preliminary step to investigate mechanisms of C. jejuni colonization in poultry in the presence of probiotics. In addition, in vivo trials to evaluate the effect of probiotics on Campylobacter colonization are conducted, taking into account the complexity introduced by the host, the feed, and the microbiota. However, the heterogeneity of the protocols used and the short time duration of the experiments lead to results that are difficult to compare and draw conclusions at the slaughter-age of broilers. Nevertheless, the combined approach using complementary in vitro and in vivo tools (cell cultures and animal experiments) leads to a better characterization of probiotic strains and could be employed to assess reduced Campylobacter spp. colonization in chickens if some parameters are optimized.
Collapse
Affiliation(s)
| | - Muriel Guyard-Nicodème
- Hygiene and Quality of Poultry and Pork Products Unit, Ploufragan/Plouzané Laboratory, ANSES, Université Bretagne LoirePloufragan, France
| | - Soumaya Messaoudi
- SECALIM Unit UMR1014, Oniris, INRA, Université Bretagne LoireNantes, France
| | - Marianne Chemaly
- Hygiene and Quality of Poultry and Pork Products Unit, Ploufragan/Plouzané Laboratory, ANSES, Université Bretagne LoirePloufragan, France
| | | | - Xavier Dousset
- SECALIM Unit UMR1014, Oniris, INRA, Université Bretagne LoireNantes, France
| | - Nabila Haddad
- SECALIM Unit UMR1014, Oniris, INRA, Université Bretagne LoireNantes, France
| |
Collapse
|
7
|
Nogueira AT, Pedrosa AT, Carabeo RA. Manipulation of the Host Cell Cytoskeleton by Chlamydia. Curr Top Microbiol Immunol 2016; 412:59-80. [PMID: 27197645 DOI: 10.1007/82_2016_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Chlamydiae are obligate intracellular pathogens. They undergo a biphasic developmental cycle differentiating between the infectious but metabolically quiescent elementary body and the vegetative, but non-infectious reticulate body. Chlamydia spends a significant portion of its development in the non-infectious stage, demanding an effective strategy of manipulating the host cells to ensure its intracellular survival and replication. A common target of all Chlamydia species studied so far is the host cell cytoskeleton, with past and recent findings revealing crucial roles in invasion, inclusion maintenance, nutrient acquisition, and egress. The molecular details of how Chlamydia co-opts the cytoskeleton is becoming clearer, with bacterial factors and their corresponding host cell targets identified.
Collapse
Affiliation(s)
- Ana T Nogueira
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Antonio T Pedrosa
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Rey A Carabeo
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| |
Collapse
|
8
|
Shyaka A, Kusumoto A, Chaisowwong W, Okouchi Y, Fukumoto S, Yoshimura A, Kawamoto K. Virulence characterization of Campylobacter jejuni isolated from resident wild birds in Tokachi area, Japan. J Vet Med Sci 2015; 77:967-72. [PMID: 25843040 PMCID: PMC4565820 DOI: 10.1292/jvms.15-0090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The prevalence of Campylobacter jejuni in wild birds is a potential hazard for human and animal health. The aim of this study was to establish the prevalence of C. jejuni in wild birds in Tokachi area, Hokkaido, Japan and investigate their virulence in vitro. In total, 173 cloacal swabs from individual wild birds were collected for the detection of Campylobacter spp. Thirty four samples (19.7%) were positive for Campylobacter of which 94.1% (32/34 samples) were C. jejuni. Additionally, one C. coli and one C. fetus were isolated. Seven C. jejuni isolates (one from crows and the other from pigeons) had important virulence genes including all three CDT genes (cdtA, cdtB and cdtC) and flaA, flaB, ciaB and cadF,
and the other isolates were lacking cdtA gene. Further studies on in vitro virulence-associated phenotypes, such as motility assay on soft agar and invasion assay in Caco-2 cells, were performed. The wild bird C. jejuni isolates adhered and invaded human cells. Although the numbers of viable intracellular bacteria of wild bird isolates were lower than a type strain NCTC11168, they persisted at 48-hr and underwent replication in host cells.
Collapse
Affiliation(s)
- Anselme Shyaka
- Section of Food Microbiology and Immunology, Diagnostic Center for Animal Health and Food Safety, Obihiro University of Agriculture and Veterinary Medicine, 2-11 Inada, Obihiro, Hokkaido 080-8555, Japan
| | | | | | | | | | | | | |
Collapse
|
9
|
Wong A, Lange D, Houle S, Arbatsky NP, Valvano MA, Knirel YA, Dozois CM, Creuzenet C. Role of capsular modified heptose in the virulence ofCampylobacter jejuni. Mol Microbiol 2015; 96:1136-58. [DOI: 10.1111/mmi.12995] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2015] [Indexed: 11/30/2022]
Affiliation(s)
- Anthony Wong
- Department of Microbiology and Immunology; Western University; DSB 3031 London ON N6A 5C1 Canada
| | - Dirk Lange
- Department of Microbiology and Immunology; Western University; DSB 3031 London ON N6A 5C1 Canada
| | | | - Nikolay P. Arbatsky
- N.D. Zelinsky Institute of Organic Chemistry; Russian Academy of Sciences; Moscow Russia
| | - Miguel A. Valvano
- Department of Microbiology and Immunology; Western University; DSB 3031 London ON N6A 5C1 Canada
- Centre for Infection and Immunity; Queen's University of Belfast; Belfast UK
| | - Yuriy A. Knirel
- N.D. Zelinsky Institute of Organic Chemistry; Russian Academy of Sciences; Moscow Russia
| | | | - Carole Creuzenet
- Department of Microbiology and Immunology; Western University; DSB 3031 London ON N6A 5C1 Canada
| |
Collapse
|
10
|
Cheap extraction of antibacterial compounds of berry pomace and their mode of action against the pathogen Campylobacter jejuni. Food Control 2014. [DOI: 10.1016/j.foodcont.2014.05.026] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
Salaheen S, White B, Bequette BJ, Biswas D. Peanut fractions boost the growth of Lactobacillus casei that alters the interactions between Campylobacter jejuni and host epithelial cells. Food Res Int 2014. [DOI: 10.1016/j.foodres.2014.05.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
12
|
Yang JW, Kim SH, Lee WW, Kim YH. Prevalence of virulence-associated genes and antimicrobial resistance of Campylobacter jejuni from ducks in Gyeongnam Province, Korea. ACTA ACUST UNITED AC 2014. [DOI: 10.7853/kjvs.2014.37.2.85] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
13
|
Salaheen S, Almario J, Biswas D. Inhibition of growth and alteration of host cell interactions of Pasteurella multocida with natural byproducts. Poult Sci 2014; 93:1375-82. [DOI: 10.3382/ps.2013-03828] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
14
|
Prevalence of virulence and cytolethal distending toxin (CDT) genes in thermophilic Campylobacter spp. from dogs and humans in Gyeongnam and Busan, Korea. ACTA ACUST UNITED AC 2014. [DOI: 10.14405/kjvr.2014.54.1.39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
15
|
Biswas D, Niwa H, Itoh K. Infection withCampylobacter jejuniInduces Tyrosine-Phosphorylated Proteins into INT-407 Cells. Microbiol Immunol 2013; 48:221-8. [PMID: 15107531 DOI: 10.1111/j.1348-0421.2004.tb03509.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The mechanisms used by Campylobacter jejuni to induce internalization into host intestinal epithelial cells have not been defined. In this study, we obtained evidence that exposure of INT-407 cells to protein kinase inhibitors results in decreased invasion of these cells by C. jejuni in a dose dependent manner. Preincubation of INT-407 cells in the presence of staurosporine, tyrphostin 46 and genistein decreased invasion of these cells by C. jejuni significantly. Moreover, C. jejuni infection of INT-407 cells induced tyrosine phosphorylation of several Triton X-100 soluble proteins with approximate molecular weights of 170, 145, 90, 60 and 55 kDa that were absent or reduced in the presence of genistein in cells after 1 hr of pretreatment. These data suggest that tyrosine protein kinase-linked pathways strongly regulate the internalization of C. jejuni into intestinal epithelial cells.
Collapse
Affiliation(s)
- Debabrata Biswas
- Laboratory of Veterinary Public Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | | | | |
Collapse
|
16
|
Molecular methods to investigate adhesion, transmigration, invasion and intracellular survival of the foodborne pathogen Campylobacter jejuni. J Microbiol Methods 2013; 95:8-23. [DOI: 10.1016/j.mimet.2013.06.031] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 06/18/2013] [Accepted: 06/28/2013] [Indexed: 01/08/2023]
|
17
|
O Cróinín T, Backert S. Host epithelial cell invasion by Campylobacter jejuni: trigger or zipper mechanism? Front Cell Infect Microbiol 2012; 2:25. [PMID: 22919617 PMCID: PMC3417527 DOI: 10.3389/fcimb.2012.00025] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 02/17/2012] [Indexed: 11/18/2022] Open
Abstract
Campylobacter jejuni, a spiral-shaped Gram-negative pathogen, is a highly frequent cause of gastrointestinal foodborne illness in humans worldwide. Clinical outcome of C. jejuni infections ranges from mild to severe diarrheal disease, and some other complications including reactive arthritis and Guillain–Barré syndrome. This review article highlights various C. jejuni pathogenicity factors, host cell determinants, and proposed signaling mechanisms involved in human host cell invasion and their potential role in the development of C. jejuni-mediated disease. A model is presented which outlines the various important interactions of C. jejuni with the intestinal epithelium, and we discuss the pro’s and con’s for the “zipper” over the “trigger” mechanism of invasion. Future work should clarify the contradictory role of some previously identified factors, and should identify and characterize novel virulence determinants, which are crucial to provide fresh insights into the diversity of strategies employed by this pathogen to cause disease.
Collapse
Affiliation(s)
- Tadhg O Cróinín
- UCD School of Biomolecular and Biomedical Sciences, University College Dublin Dublin, Ireland
| | | |
Collapse
|
18
|
Krause-Gruszczynska M, Boehm M, Rohde M, Tegtmeyer N, Takahashi S, Buday L, Oyarzabal OA, Backert S. The signaling pathway of Campylobacter jejuni-induced Cdc42 activation: Role of fibronectin, integrin beta1, tyrosine kinases and guanine exchange factor Vav2. Cell Commun Signal 2011; 9:32. [PMID: 22204307 PMCID: PMC3286397 DOI: 10.1186/1478-811x-9-32] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 12/28/2011] [Indexed: 11/12/2022] Open
Abstract
Background Host cell invasion by the foodborne pathogen Campylobacter jejuni is considered as one of the primary reasons of gut tissue damage, however, mechanisms and key factors involved in this process are widely unclear. It was reported that small Rho GTPases, including Cdc42, are activated and play a role during invasion, but the involved signaling cascades remained unknown. Here we utilised knockout cell lines derived from fibronectin-/-, integrin-beta1-/-, focal adhesion kinase (FAK)-/- and Src/Yes/Fyn-/- deficient mice, and wild-type control cells, to investigate C. jejuni-induced mechanisms leading to Cdc42 activation and bacterial uptake. Results Using high-resolution scanning electron microscopy, GTPase pulldowns, G-Lisa and gentamicin protection assays we found that each studied host factor is necessary for induction of Cdc42-GTP and efficient invasion. Interestingly, filopodia formation and associated membrane dynamics linked to invasion were only seen during infection of wild-type but not in knockout cells. Infection of cells stably expressing integrin-beta1 variants with well-known defects in fibronectin fibril formation or FAK signaling also exhibited severe deficiencies in Cdc42 activation and bacterial invasion. We further demonstrated that infection of wild-type cells induces increasing amounts of phosphorylated FAK and growth factor receptors (EGFR and PDGFR) during the course of infection, correlating with accumulating Cdc42-GTP levels and C. jejuni invasion over time. In studies using pharmacological inhibitors, silencing RNA (siRNA) and dominant-negative expression constructs, EGFR, PDGFR and PI3-kinase appeared to represent other crucial components upstream of Cdc42 and invasion. siRNA and the use of Vav1/2-/- knockout cells further showed that the guanine exchange factor Vav2 is required for Cdc42 activation and maximal bacterial invasion. Overexpression of certain mutant constructs indicated that Vav2 is a linker molecule between Cdc42 and activated EGFR/PDGFR/PI3-kinase. Using C. jejuni mutant strains we further demonstrated that the fibronectin-binding protein CadF and intact flagella are involved in Cdc42-GTP induction, indicating that the bacteria may directly target the fibronectin/integrin complex for inducing signaling leading to its host cell entry. Conclusion Collectively, our findings led us propose that C. jejuni infection triggers a novel fibronectin→integrin-beta1→FAK/Src→EGFR/PDGFR→PI3-kinase→Vav2 signaling cascade, which plays a crucial role for Cdc42 GTPase activity associated with filopodia formation and enhances bacterial invasion.
Collapse
|
19
|
Richardson LJ, Cox NA, Buhr RJ, Harrison MA. Isolation of Campylobacter from Circulating Blood of Commercial Broilers. Avian Dis 2011; 55:375-8. [DOI: 10.1637/9613-121310-reg.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Takenouchi S, Sugahara T. Lactate dehydrogenase enhances immunoglobulin production by human hybridoma and human peripheral blood lymphocytes. Cytotechnology 2011; 42:133-43. [PMID: 19002935 DOI: 10.1023/b:cyto.0000015838.06536.de] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lactate dehydrogenase (LDH) derived from rabbit muscle enhanced IgM production by human-human hybridoma HB4C5 cells 12.4-fold at 320 mug/ml under serum-free conditions. LDHs from pig muscle and pig heart also accelerated IgM production 8.4- and 6.4-fold, respectively. The immunoglobulin production stimulating activity of LDH was not accompanied by activation of cell proliferation. LDH from rabbit muscle facilitated IgM and IgG production by human peripheral blood lymphocytes. This means LDH stimulates immunoglobulin production not only by the specified hybridoma cell line, but also by unspecified immunoglobulin producers. LDH from rabbit muscle enhanced IgM production of transcription-suppressed HB4C5 cells treated with actinomycin D. The immunoglobulin production-stimulating factors (IPSFs) effect of LDH was slightly weakened by sodium fluoride (translation inhibitor) treatment of HB4C5. Moreover, the amount of intracellular IgM of monensin-treated HB4C5 cells was obviously enhanced by LDH. This result means that the IPSF effect of LDH is irrelevant to the post-translation activity of target cells. It is expected from these findings that LDH from rabbit muscle accelerates the translation step to enhance immunoglobulin productivity. The immunoglobulin production-stimulating activity of LDH was inhibited by colchicine, endocytosis inhibitor. This fact suggests that it is necessary for LDH to be taken by target cells to act as an IPSF.
Collapse
Affiliation(s)
- Satoshi Takenouchi
- Faculty of Agriculture, Ehime University, 3-5-7 Tarumi, Matsuyama, Ehime, 790-8566, Japan
| | | |
Collapse
|
21
|
Campylobacter fetus translocation across Caco-2 cell monolayers. Microb Pathog 2010; 49:260-72. [PMID: 20600794 DOI: 10.1016/j.micpath.2010.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 04/23/2010] [Accepted: 06/16/2010] [Indexed: 12/26/2022]
Abstract
Campylobacter fetus is a recognized pathogen of cattle and sheep, though human infection has also been reported. Ingestion of contaminated food or water is a proposed route of transmission for both humans and animals. The subsequent detection of the organism from extra-intestinal and systemic locations implies an ability to translocate across epithelial barriers. To determine how C. fetus disseminates from the intestine, Caco-2 cells cultured on porous membrane supports, were used as model intestinal epithelial cell monolayers. C. fetus was found to translocate equally well in both apical-to-basolateral and basolateral-to-apical directions for up to 24 h without altering Caco-2 cell monolayer permeability as assessed by transepithelial resistance and absence of paracellular diffusion of FITC-inulin. Using modified antibiotic protection assays, C. fetus was also observed to invade and subsequently egress from Caco-2 cells. Caco-2 cell invasion and translocation occurred independently of C. fetus S layer expression. Scanning and transmission electron microscopy revealed the presence of C. fetus associated with both apical and basal surfaces as well as in intracellular locations. C. fetus was, however, never observed in paracellular locations nor associated with Caco-2 cells junctions. Neither C. fetus invasion nor translocation across Caco-2 cell monolayers was impacted by latrunculin A, though translocation was enhanced in the presence of cytochalasin D which disrupted tight junctions. Tubulin cytoskeleton disrupting agents, colchicine and vinblastine, did inhibit C. fetus translocation though entry into Caco-2 cells remained unaffected. Together, translocation without disrupting monolayer integrity, invasion and egression from Caco-2 cells, electron microscopy observations and the requirement of a functional tubulin cytoskeleton for translocation, support a transcellular mechanism of C. fetus translocation across Caco-2 cell monolayers. The ability to invade and subsequently egress would contribute to establishment of an infecting C. fetus population in the host, while the demonstrated ability to translocate across model intestinal epithelial barriers accounts for the observed in vivo recovery of C. fetus from extra-intestinal locations.
Collapse
|
22
|
van Putten JPM, van Alphen LB, Wösten MMSM, de Zoete MR. Molecular mechanisms of campylobacter infection. Curr Top Microbiol Immunol 2010; 337:197-229. [PMID: 19812984 DOI: 10.1007/978-3-642-01846-6_7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Campylobacter jejuni is the principal bacterial foodborne pathogen. A major challenge still is to identify the virulence strategies exploited by C. jejuni. Recent genomics, proteomics, and metabolomics approaches indicate that C. jejuni displays extensive inter- and intrastrain variation. The diverse behavior enables bacterial adaptation to different environmental conditions and directs interactions with the gut mucosa. Here, we report recent progress in understanding the molecular mechanisms and functional consequences of the phenotype diversity. The results suggest that C. jejuni actively penetrates the intestinal mucus layer, secretes proteins mainly via its flagellar apparatus, is engulfed by intestinal cells, and can disrupt the integrity of the epithelial lining. C. jejuni stimulates the proinflammatory pathway and the production of a large repertoire of cytokines, chemokines, and innate effector molecules. Novel experimental infection models suggest that the activation of the innate immune response is important for the development of intestinal pathology.
Collapse
Affiliation(s)
- Jos P M van Putten
- Department of Infectious Diseases & Immunology, Utrecht University, Yalelaan 1, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
23
|
Cox NA, Richardson LJ, Buhr RJ, Fedorka-Cray PJ. Campylobacter species occurrence within internal organs and tissues of commercial caged Leghorn laying hens. Poult Sci 2009; 88:2449-56. [PMID: 19834099 DOI: 10.3382/ps.2009-00195] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Campylobacter spp. are frequently present in the intestinal tract and internal tissues of broiler breeder and broiler chickens. Campylobacter spp. ecology in commercial Leghorn laying hens has not been extensively studied. The objectives of the current study were to determine 1) Campylobacter spp. presence in the reproductive tract, lymphoid organs, liver-gallbladder, and ceca of commercial Leghorn laying hens; 2) species of Campylobacter present; and 3) antimicrobial resistance pattern of Campylobacter isolates. In study 1, three flocks ranging from 94 to 105 wk of age were sampled from a commercial laying complex. In study 2, two flocks, 82 and 84 wk of age, were sampled from a separate complex. Hens were killed, defeathered, aseptically necropsied, and the spleen, liver-gallbladder, ovarian follicles, and upper (infundibulum, magnum, and isthmus) and lower (shell gland and vagina) reproductive tracts were aseptically removed before the ceca. Samples were packed on ice and transported to the laboratory for evaluation. For speciation, a standard BAX real-time PCR method was used while susceptibility testing was performed using US National Antimicrobial Resistance Monitoring System (NARMS) standards and recommended quality control organisms. Isolates were examined for susceptibility using a semi-automated testing system (Sensititer) to the following 9 antimicrobials: azithromycin, clindamycin, ciprofloxacin, erythromycin, florfenicol, gentamicin, nalidixic acid, telithromycin, and tetracycline. In study 1, the isolation rate was 13, 67, 53, 3, 13, and 57% from the ovarian follicles, lower reproductive tract, upper reproductive tract, spleen, liver-gallbladder, and ceca, respectively. In study 2, the isolation rate was 17, 43, 33, 20, 17, and 73% from the ovarian follicles, lower reproductive tract, upper reproductive tract, spleen, liver-gallbladder, and ceca, respectively. Overall, 50% of isolates were Campylobacter jejuni, 49% Campylobacter coli, and 1% Campylobacter lari. In study 1, all of the isolates were pan-susceptible. In study 2, thirty-seven percent of the isolates were resistant to tetracycline. Commercial table egg laying hens housed in colony cages on wire floors had diverse Campylobacter spp. recovered from different tissues and these isolates were not resistant to a broad range of antimicrobials.
Collapse
Affiliation(s)
- N A Cox
- USDA, Agricultural Research Service, Poultry Microbiological Safety Research Unit, GA, USA.
| | | | | | | |
Collapse
|
24
|
Dasti JI, Tareen AM, Lugert R, Zautner AE, Gross U. Campylobacter jejuni: a brief overview on pathogenicity-associated factors and disease-mediating mechanisms. Int J Med Microbiol 2009; 300:205-11. [PMID: 19665925 DOI: 10.1016/j.ijmm.2009.07.002] [Citation(s) in RCA: 269] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 06/17/2009] [Accepted: 07/04/2009] [Indexed: 12/11/2022] Open
Abstract
Campylobacter jejuni has long been recognized as a cause of bacterial food-borne illness, and surprisingly, it remains the most prevalent bacterial food-borne pathogen in the industrial world to date. Natural reservoirs for this Gram-negative, spiral-shaped bacterium are wild birds, whose intestines offer a suitable biological niche for the survival and dissemination of C. jejuni Chickens become colonized shortly after birth and are the most important source for human infection. In the last decade, effective intervention strategies to limit infections caused by this elusive pathogen were hindered mainly because of a paucity in understanding the virulence mechanisms of C. jejuni and in part, unavailability of an adequate animal model for the disease. However, recent developments in deciphering molecular mechanisms of virulence of C. jejuni made it clear that C. jejuni is a unique pathogen, being able to execute N-linked glycosylation of more than 30 proteins related to colonization, adherence, and invasion. Moreover, the flagellum is not only depicted to facilitate motility but as well secretion of Campylobacter invasive antigens (Cia). The only toxin of C. jejuni, the so-called cytolethal distending toxin (CdtA,B,C), seems to be important for cell cycle control and induction of host cell apoptosis and has been recognized as a major pathogenicity-associated factor. In contrast to other diarrhoea-causing bacteria, no other classical virulence factors have yet been identified in C. jejuni. Instead, host factors seem to play a major role for pathogenesis of campylobacteriosis of man. Indeed, several lines of evidence suggest exploitation of different adaptation strategies by this pathogen depending on its requirement, whether to establish itself in the natural avian reservoir or during the course of human infection.
Collapse
Affiliation(s)
- Javid I Dasti
- Institute of Medical Microbiology, University Medical Centre Göttingen, Kreuzbergring 57, D-37075 Göttingen, Germany
| | | | | | | | | |
Collapse
|
25
|
Verhoeff-Bakkenes L, Hazeleger W, de Jonge R, Zwietering M. Campylobacter jejuni: a study on environmental conditions affecting culturability andin vitroadhesion/invasion. J Appl Microbiol 2009; 106:924-31. [DOI: 10.1111/j.1365-2672.2008.04072.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Kalischuk LD, Inglis GD, Buret AG. Campylobacter jejuni induces transcellular translocation of commensal bacteria via lipid rafts. Gut Pathog 2009; 1:2. [PMID: 19338680 PMCID: PMC2653720 DOI: 10.1186/1757-4749-1-2] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Accepted: 02/03/2009] [Indexed: 02/08/2023] Open
Abstract
Background Campylobacter enteritis represents a risk factor for the development of inflammatory bowel disease (IBD) via unknown mechanisms. As IBD patients exhibit inflammatory responses to their commensal intestinal microflora, factors that induce translocation of commensal bacteria across the intestinal epithelium may contribute to IBD pathogenesis. This study sought to determine whether Campylobacter induces translocation of non-invasive intestinal bacteria, and characterize underlying mechanisms. Methods Mice were infected with C. jejuni and translocation of intestinal bacteria was assessed by quantitative bacterial culture of mesenteric lymph nodes (MLNs), liver, and spleen. To examine mechanisms of Campylobacter-induced bacterial translocation, transwell-grown T84 monolayers were inoculated with non-invasive Escherichia coli HB101 ± wild-type Campylobacter or invasion-defective mutants, and bacterial internalization and translocation were measured. Epithelial permeability was assessed by measuring flux of a 3 kDa dextran probe. The role of lipid rafts was assessed by cholesterol depletion and caveolin co-localization. Results C. jejuni 81–176 induced translocation of commensal intestinal bacteria to the MLNs, liver, and spleen of infected mice. In T84 monolayers, Campylobacter-induced internalization and translocation of E. coli occurred via a transcellular pathway, without increasing epithelial permeability, and was blocked by depletion of epithelial plasma membrane cholesterol. Invasion-defective mutants and Campylobacter-conditioned cell culture medium also induced E. coli translocation, indicating that C. jejuni does not directly 'shuttle' bacteria into enterocytes. In C. jejuni-treated monolayers, translocating E. coli associated with lipid rafts, and this phenomenon was blocked by cholesterol depletion. Conclusion Campylobacter, regardless of its own invasiveness, promotes the translocation of non-invasive bacteria across the intestinal epithelium via a lipid raft-mediated transcellular process.
Collapse
Affiliation(s)
- Lisa D Kalischuk
- Department of Biological Sciences, Inflammation Research Network, University of Calgary, Calgary, AB, Canada.
| | | | | |
Collapse
|
27
|
CHANSIRIPORNCHAI N, SASIPREEYAJAN J. PCR Detection of Four Virulence-Associated Genes of Campylobacter jejuni Isolates from Thai Broilers and Their Abilities of Adhesion to and Invasion of INT-407 Cells. J Vet Med Sci 2009; 71:839-44. [DOI: 10.1292/jvms.71.839] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Niwat CHANSIRIPORNCHAI
- Department of Veterinary Medicine, Faculty of Veterinary Sciences, Chulalongkorn University
| | - Jiroj SASIPREEYAJAN
- Department of Veterinary Medicine, Faculty of Veterinary Sciences, Chulalongkorn University
| |
Collapse
|
28
|
Enhanced microscopic definition of Campylobacter jejuni 81-176 adherence to, invasion of, translocation across, and exocytosis from polarized human intestinal Caco-2 cells. Infect Immun 2008; 76:5294-304. [PMID: 18765731 DOI: 10.1128/iai.01408-07] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Campylobacter jejuni-mediated pathogenesis involves gut adherence and translocation across intestinal cells. The current study was undertaken to examine the C. jejuni interaction with and translocation across differentiated Caco-2 cells to better understand Campylobacter's pathogenesis. The efficiency of C. jejuni 81-176 invasion of Caco-2 cells was two- to threefold less than the efficiency of invasion of INT407 cells. Adherence-invasion analyses indicated that C. jejuni 81-176 adhered to most INT407 cells but invaded only about two-thirds of the host cells over 2 h (two bacteria/cell). In contrast, only 11 to 17% of differentiated Caco-2 cells were observed to bind and internalize either C. jejuni strain 81-176 or NCTC 11168, and a small percentage of infected Caco-2 cells contained 5 to 20 internalized bacteria per cell after 2 h. Electron microscopy revealed that individual C. jejuni cells adhered to the tips of host cell microvilli via intimate flagellar contacts and by lateral bacterial binding to the sides of microvilli. Next, bacteria were observed to bind at the apical host membrane surface via presumed interactions at one pole of the bacterium and with host membrane protrusions located near intercellular junctions. The latter contacts apparently resulted in coordinated, localized plasma membrane invagination, causing simultaneous internalization of bacteria into an endosome. Passage of this Campylobacter endosome intracellularly from the apical surface to the basolateral surface occurred over time, and bacterial release apparently resulted from endosome-basolateral membrane fusion (i.e., exocytosis). Bacteria were found intercellularly below tight junctions at 60 min postinfection, but not at earlier times. This study revealed unique host cell adherence contacts, early endocytosis-specific structures, and a presumptive exocytosis component of the transcellular transcytosis route.
Collapse
|
29
|
Fearnley C, Manning G, Bagnall M, Javed MA, Wassenaar TM, Newell DG. Identification of hyperinvasive Campylobacter jejuni strains isolated from poultry and human clinical sources. J Med Microbiol 2008; 57:570-580. [PMID: 18436589 DOI: 10.1099/jmm.0.47803-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Campylobacter jejuni causes gastroenteritis with a variety of symptoms in humans. In the absence of a suitable animal model, in vitro models have been used to study virulence traits such as invasion and toxin production. In this study, 113 C. jejuni isolates from poultry and poultry-related (n=74) environments as well as isolates from human cases (n=39) of campylobacteriosis and bacteraemia were tested for invasiveness using INT 407 cells. The method was sufficiently reproducible to observe a spectrum of invasiveness amongst strains. As a result, strains were classified as low, high and hyper-invasive. The majority of strains (poultry and human) were low invaders (82 % and 88 %, respectively). High invasion was found for 5 % of human strains and 11 % of poultry-related isolates. However, only 1 % of poultry strains were classified as hyperinvasive compared to 13 % of human isolates (P=0.0182). Of those isolates derived from the blood of bacteraemic patients, 20 % were hyperinvasive, though this correlation was not statistically significant. An attempt was made to correlate invasiveness with the presence of seven genes previously reported to be associated with virulence. Most of these genes did not correlate with invasiveness, but gene cj0486 was weakly over-represented, and a negative correlation was observed for the gene ciaB. This trend was stronger when the two genes were analysed together, thus ciaB(-) cj0486(+) was over-represented in high and hyperinvasive strains, with low invaders more commonly found to lack these genes (P=0.0064).
Collapse
Affiliation(s)
| | - Georgina Manning
- School of Science and Technology, Nottingham Trent University, NG11 8NS, UK
| | - Mary Bagnall
- Veterinary Laboratories Agency (Weybridge), Surrey KT15 3NB, UK
| | | | - Trudy M Wassenaar
- Molecular Microbiology and Genomics Consultants, Zotzenheim, Germany
| | - Diane G Newell
- Veterinary Laboratories Agency (Weybridge), Surrey KT15 3NB, UK
| |
Collapse
|
30
|
van Alphen LB, Bleumink-Pluym NMC, Rochat KD, van Balkom BWM, Wösten MMSM, van Putten JPM. Active migration into the subcellular space precedes Campylobacter jejuni invasion of epithelial cells. Cell Microbiol 2008; 10:53-66. [PMID: 18052944 DOI: 10.1111/j.1462-5822.2007.01014.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The bacterial pathogen Campylobacter jejuni invades mucosal cells via largely undefined and rather inefficient (0.01-2 bacteria per cell) mechanisms. Here we report a novel, highly efficient C. jejuni infection pathway resulting in 10-15 intracellular bacteria per cell within 3 h of infection. Electron microscopy, pulse-chase infection assays and time-lapse multiphoton laser confocal microscopy demonstrated that the mechanism involved active and rapid migration of the pathogen into the subcellular space (termed 'subvasion'), followed by bacterial entry ('invasion') at the cell basis. Efficient subvasion was maximal after repeated rounds of selection for the subvasive phenotype. Targeted mutagenesis indicated that the CadF, JlpA or PEB1 adhesins were not required. Dissection of the selected and parental phenotypes by SDS-PAGE yielded comparable capsule polysaccharide and lipooligosaccharide profiles. Proteomics revealed reduced amounts of the chemotaxis protein CheW for the subvasive phenotype. Swarming assays confirmed that the selected phenotype exhibited altered migration behaviour. Introduction of a plasmid carrying chemotaxis genes into the subvasive strain yielded wild-type subvasion levels and migration behaviour. These results indicate that alterations in the bacterial migration machinery enable C. jejuni to actively penetrate the subcellular space and gain access to the cell interior with unprecedented efficiency.
Collapse
Affiliation(s)
- Lieke B van Alphen
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
31
|
Lack of response of INT-407 cells to the presence of non-culturable Campylobacter jejuni. Epidemiol Infect 2007; 136:1401-6. [PMID: 18081950 DOI: 10.1017/s0950268807000040] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Many contradictory articles on the infectivity of non-culturable Campylobacter jejuni can be found. We studied the effect of non-culturable C. jejuni in an in vitro assay. To prevent the potential effect of a few culturable bacteria in the non-culturable suspension, INT-407 cells, which mimic the outer cell layer in the small intestines, were exposed to culturable C. jejuni suspensions with or without non-culturable C. jejuni. The number of bacteria adhering to and/or invading INT-407 cells and the IL-8 secretion were measured. No differences were found between bacterial suspensions with or without non-culturable C. jejuni added. These findings show that non-culturable C. jejuni do not adhere to or invade INT-407 cells and do not induce an immune response. As previous studies showed a correlation between the used in vitro assays and the effect in vivo, our study strongly suggests that culturability is a good indicator of the risk for C. jejuni infection.
Collapse
|
32
|
Abstract
Campylobacter jejuni is a foodborne bacterial pathogen that is common in the developed world. However, we know less about its biology and pathogenicity than we do about other less prevalent pathogens. Interest in C. jejuni has increased in recent years as a result of the growing appreciation of its importance as a pathogen and the availability of new model systems and genetic and genomic technologies. C. jejuni establishes persistent, benign infections in chickens and is rapidly cleared by many strains of laboratory mouse, but causes significant inflammation and enteritis in humans. Comparing the different host responses to C. jejuni colonization should increase our understanding of this organism.
Collapse
Affiliation(s)
- Kathryn T Young
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
33
|
Krause-Gruszczynska M, Rohde M, Hartig R, Genth H, Schmidt G, Keo T, König W, Miller WG, Konkel ME, Backert S. Role of the small Rho GTPases Rac1 and Cdc42 in host cell invasion of Campylobacter jejuni. Cell Microbiol 2007; 9:2431-44. [PMID: 17521326 DOI: 10.1111/j.1462-5822.2007.00971.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Host cell invasion of the food-borne pathogen Campylobacter jejuni is one of the primary reasons of tissue damage in humans but molecular mechanisms are widely unclear. Here, we show that C. jejuni triggers membrane ruffling in the eukaryotic cell followed by invasion in a very specific manner first with its tip followed by the flagellar end. To pinpoint important signalling events involved in the C. jejuni invasion process, we examined the role of small Rho family GTPases. Using specific GTPase-modifying toxins, inhibitors and GTPase expression constructs we show that Rac1 and Cdc42, but not RhoA, are involved in C. jejuni invasion. In agreement with these observations, we found that internalization of C. jejuni is accompanied by a time-dependent activation of both Rac1 and Cdc42. Finally, we show that the activation of these GTPases involves different host cell kinases and the bacterial fibronectin-binding protein CadF. Thus, CadF is a bifunctional protein which triggers bacterial binding to host cells as well as signalling leading to GTPase activation. Collectively, our results suggest that C. jejuni invade host target cells by a unique mechanism and the activation of the Rho GTPase members Rac1 and Cdc42 plays a crucial role in this entry process.
Collapse
Affiliation(s)
- Malgorzata Krause-Gruszczynska
- Department of Medical Microbiology, Otto von Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Byrne CM, Clyne M, Bourke B. Campylobacter jejuni adhere to and invade chicken intestinal epithelial cells in vitro. Microbiology (Reading) 2007; 153:561-569. [PMID: 17259628 DOI: 10.1099/mic.0.2006/000711-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Campylobacter jejuni is a common cause of serious diarrhoeal disease in humans, in contrast to the avian population, where exposure results in prolonged colonization at high density without disease. Colonized poultry present a significant source of infection to humans worldwide. The aim of this work was to compare the interaction of Campylobacter with primary intestinal cells from humans and poultry to identify factors that account for the divergent outcome following Campylobacter exposure. A primary intestinal cell model of Campylobacter infection was developed using cells grown from human and chicken intestinal biopsies. The cultured cells were infected with a number of strains of Campylobacter. Invasion by C. jejuni and the influence of intestinal mucus on Campylobacter internalization were studied by fluorescence microscopy and gentamicin protection assays. C. jejuni invaded primary human intestinal cells in a microtubule-, microfilament- and caveolin-dependent manner. Entry of C. jejuni into primary chicken intestinal cells also occurred. Chicken mucus, but not intestinal mucus of human origin, significantly reduced infection of primary human intestinal cells. Avian mucus appears to inhibit Campylobacter from interacting with epithelial cell surfaces.
Collapse
Affiliation(s)
- Catherine M Byrne
- The Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin 12, and The Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science, University College Dublin, Ireland
| | - Marguerite Clyne
- The Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin 12, and The Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science, University College Dublin, Ireland
| | - Billy Bourke
- The Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin 12, and The Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science, University College Dublin, Ireland
| |
Collapse
|
35
|
Biswas D, Fernando U, Reiman C, Willson P, Potter A, Allan B. Effect of Cytolethal Distending Toxin of Campylobacter jejuni on Adhesion and Internalization in Cultured Cells and in Colonization of the Chicken Gut. Avian Dis 2006; 50:586-93. [PMID: 17274298 DOI: 10.1637/7514-020706r1.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Campylobacter jejuni produces cytolethal distending toxin (CDT) that causes host cells to arrest during their cell cycle and that is involved in the pathogenesis of inflammatory diarrhea in humans. To assess the role of CDT in adherence and invasion of different cultured host cells (HeLa and HD-11) and in colonization of the chicken intestine, the genes of C jejuni NCTC11168 encoding the toxin subunits (cdtA, cdtB, and cdtC) were inactivated by insertional mutagenesis. No significant difference was found in adhesion of the wild-type C. jejuni and the isogenic mutants to HeLa and HD-11 cells. All of the mutants exhibited a decrease (>10-fold) in the ability to invade HeLa cells, but no significant difference was noticed for HD-11 cells. The ability of mutants to colonize birds either directly or by horizontal transfer was unchanged. These data indicated that although the production of cytotoxin does not play a role in the adherence to either human or avian cells, it may play a role in the invasion, survival, or both of C. jejuni in human cells, which are more susceptible to C. jejuni internalization. The CDT also does not seem to play a role in the colonization of poultry.
Collapse
Affiliation(s)
- Debabrata Biswas
- Vaccine and Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Road, Saskatoon, Saskatchewan, Canada S7N5E3
| | | | | | | | | | | |
Collapse
|
36
|
Johanesen PA, Dwinell MB. Flagellin-independent regulation of chemokine host defense in Campylobacter jejuni-infected intestinal epithelium. Infect Immun 2006; 74:3437-47. [PMID: 16714574 PMCID: PMC1479283 DOI: 10.1128/iai.01740-05] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Campylobacter jejuni is a leading cause of bacterial food-borne diarrheal disease throughout the world and the most frequent antecedent of autoimmune neuropathy Guillain-Barré syndrome. While infection is associated with immune memory, little is known regarding the role of the epithelium in targeting dendritic cells (DC) for initiating the appropriate adaptive immune response to C. jejuni. The objective of this study was to define the role for the intestinal epithelium in the induction of the adaptive immune response in C. jejuni infection by assessing the production of DC and T-cell chemoattractants. Human T84 epithelial cells were used as model intestinal epithelia. Infection of T84 cells with C. jejuni dose- and time-dependently up-regulated DC and T-cell chemokine gene transcription and secretion. Induction required live bacteria and was in the physiologically relevant direction for attraction of mucosal immunocytes. C. jejuni-activated NF-kappaB signaling was shown to be essential for proinflammatory chemokine secretion. Notably, C. jejuni secretion occurred independently of flagellin identification by Toll-like receptor 5. Secretion of a DC chemoattractant by differing clinical C. jejuni isolates suggested adherence/invasion were key virulence determinants of epithelial chemokine secretion. The regulated epithelial expression of DC and T-cell chemoattractants suggests a mechanism for the directed trafficking of immune cells required for the initiation of adaptive immunity in campylobacteriosis. Chemokine secretion occurs despite Campylobacter evasion of the flagellin pattern recognition receptor, suggesting that alternate host defense strategies limit disease pathogenesis.
Collapse
Affiliation(s)
- Priscilla A Johanesen
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | |
Collapse
|
37
|
Hu L, McDaniel JP, Kopecko DJ. Signal transduction events involved in human epithelial cell invasion by Campylobacter jejuni 81-176. Microb Pathog 2006; 40:91-100. [PMID: 16426812 DOI: 10.1016/j.micpath.2005.11.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2005] [Revised: 11/14/2005] [Accepted: 11/14/2005] [Indexed: 11/26/2022]
Abstract
Analyses of invasive enteric bacteria (e.g. Shigella, Salmonella, Listeria, and Campylobacter) have shown that these pathogens initiate orchestrated signal transduction cascades in host cells leading to host cytoskeletal rearrangements that result in bacterial uptake. This current study was specifically aimed at examining the involvement of host membrane caveolae and certain protein kinases in epithelial cell invasion by C. jejuni strain 81-176, for which we have previously characterized the kinetics of entry and a unique microtubule-dependent mechanism of internalization. Utilizing in vitro cultured cell invasion assays with a gentamicin-kill step, disruption of membrane caveolae by pretreatment of INT407 cell monolayers with filipin III reduced C. jejuni 81-176 entry by >95%. Strain 81-176 uptake into INT407 cells was markedly inhibited by monolayer pretreatment with the protein kinase inhibitors genistein and staurosporine, or specific inhibitors of PI 3-kinase, wortmannin and LY294002. Western blot analysis using monoclonal anti-protein tyrosine phosphorylation antibody revealed distinctive changes during invasion in phosphorylation of at least nine proteins. Further inhibitor studies indicated that heterotrimeric G proteins, plus ERK and p38 MAP kinase activation are also involved in C. jejuni 81-176 invasion. These results suggest that C. jejuni 81-176 interact at host cell surface membrane caveolae with G protein-coupled receptors, which presumably trigger G-proteins and kinases to activate host proteins including PI 3-kinase and MAP kinases, that appear to be intimately involved in the events controlling 81-176 internalization.
Collapse
Affiliation(s)
- Lan Hu
- Laboratory of Enteric and Sexually Transmitted Diseases, Center for Biologics Evaluation and Research, FDA, NIH Campus Building 29/420, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
38
|
Luck SN, Bennett-Wood V, Poon R, Robins-Browne RM, Hartland EL. Invasion of epithelial cells by locus of enterocyte effacement-negative enterohemorrhagic Escherichia coli. Infect Immun 2005; 73:3063-71. [PMID: 15845514 PMCID: PMC1087320 DOI: 10.1128/iai.73.5.3063-3071.2005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The majority of enterohemorrhagic Escherichia coli (EHEC) strains associated with severe disease carry the locus of enterocyte effacement (LEE) pathogenicity island, which encodes the ability to induce attaching and effacing lesions on the host intestinal mucosa. While LEE is essential for colonization of the host in these pathogens, strains of EHEC that do not carry LEE are regularly isolated from patients with severe disease, although little is known about the way these organisms interact with the host epithelium. In this study, we compared the adherence properties of clinical isolates of LEE-negative EHEC with those of LEE-positive EHEC O157:H7. Transmission electron microscopy revealed that LEE-negative EHEC O113:H21 was internalized by Chinese hamster ovary (CHO-K1) epithelial cells and that intracellular bacteria were located within a membrane-bound vacuole. In contrast, EHEC O157:H7 remained extracellular and intimately attached to the epithelial cell surface. Quantitative gentamicin protection assays confirmed that EHEC O113:H21 was invasive and also showed that several other serogroups of LEE-negative EHEC were internalized by CHO-K1 cells. Invasion by EHEC O113:H21 was significantly reduced in the presence of the cytoskeletal inhibitors cytochalasin D and colchicine and the pan-Rho GTPase inhibitor compactin, whereas the tyrosine kinase inhibitor genistein had no significant impact on bacterial invasion. In addition, we found that EHEC O113:H21 was invasive for the human colonic cell lines HCT-8 and Caco-2. Overall these studies suggest that isolates of LEE-negative EHEC may employ a mechanism of host cell invasion to colonize the intestinal mucosa.
Collapse
Affiliation(s)
- Shelley N Luck
- Department of Microbiology, Monash University, Victoria 3800, Australia
| | | | | | | | | |
Collapse
|
39
|
Friis LM, Pin C, Pearson BM, Wells JM. In vitro cell culture methods for investigating Campylobacter invasion mechanisms. J Microbiol Methods 2005; 61:145-60. [PMID: 15722140 DOI: 10.1016/j.mimet.2004.12.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2004] [Revised: 12/04/2004] [Accepted: 12/06/2004] [Indexed: 01/24/2023]
Abstract
Studying the mechanisms of Campylobacter pathogenesis is complicated by the lack of simple animal models that mimic the disease seen in humans. In vitro cell culture methods provide a useful alternative to investigate the interactions between Campylobacter and the host epithelium that occur during infection. In the genomics era there is an increasing use of in vitro cell culture techniques to interrogate the potential role of different genes in pathogenesis. The aim of this review was to discuss the suitability and limitations of the various experimental approaches that might be adopted. We review current knowledge concerning the influence of cell-specific as well as bacterial factors required for Campylobacter invasion such as flagella and secreted proteins. The involvement and effects of phase variation on the results of invasion studies in cell culture emphasise the need to verify observed strain variations. We present the use of a mathematical Invasion Success Model to analyse Campylobacter invasion and show that it can be used to derive three strain dependent characteristics Imax, k, and I0. Even by combining data from independent experiments the Invasion Success Model can be used to statistically compare Campylobacter strains for their invasion of epithelial cells. Recommendations are given for the adoption of standard assay parameters and analytical methods such as the Invasion Success Model in order to facilitate comparison of data generated in different laboratories.
Collapse
Affiliation(s)
- L M Friis
- Institute of Food Research, Colney Lane, Norwich, NR4 7UA, United Kingdom
| | | | | | | |
Collapse
|
40
|
|
41
|
Mooney A, Byrne C, Clyne M, Johnson-Henry K, Sherman P, Bourke B. Invasion of human epithelial cells by Campylobacter upsaliensis. Cell Microbiol 2004; 5:835-47. [PMID: 14531898 DOI: 10.1046/j.1462-5822.2003.00325.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Few data exist on the interaction of Campylobacter upsaliensis with host cells, and the potential for this emerging enteropathogen to invade epithelial cells has not been explored. We have characterized the ability of C. upsaliensis to invade both cultured epithelial cell lines and primary human small intestinal cells. Epithelial cell lines of intestinal origin appeared to be more susceptible to invasion than non-intestinal-derived cells. Of three bacterial isolates studied, a human clinical isolate, CU1887, entered cells most efficiently. Although there was a trend towards more efficient invasion of Caco-2 cells by C. upsaliensis CU1887 at lower initial inocula, actual numbers of intracellular organisms increased with increasing multiplicity of infection and with prolonged incubation period. Confocal microscopy revealed C. upsaliensis within primary human small intestinal cells. Both Caco-2 and primary cells in non-confluent areas of the infected monolayers were substantially more susceptible to infection than confluent cells. The specific cytoskeletal inhibitors cytochalasin B, cytochalasin D and vinblastine attenuated invasion of Caco-2 cells in a concentration-dependent manner, providing evidence for both microtubule- and microfilament-dependent uptake of C. upsaliensis. Electron microscopy revealed the presence of organisms within Caco-2 cell cytoplasmic vacuoles. C. upsaliensis is capable of invading epithelial cells and appears to interact with host cell cytoskeletal structures in order to gain entry to the intracellular environment. Entry into cultured primary intestinal cells ex vivo provides strong support for the role of host cell invasion during human enteric C. upsaliensis infection.
Collapse
Affiliation(s)
- Adele Mooney
- Department of Paediatrics, The Conway Institute for Biomolecular and Biomedical Research, University College Dublin, The Children's Research Centre, Our Lady's Hospital for Sick Children, Crumlin, Dublin 12, Ireland
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Campylobacterial infections are the most common cause of bacterial enterocolitis in humans. Among children, especially in developing countries, Campylobacter infections can cause severe life-threatening diarrheal disease. Although usually associated with a benign outcome in the developed world, the burden of illness posed by Campylobacter infections is enormous, and serious neurologic sequelae also can occur. For a variety of reasons our understanding of the molecular and cellular pathogenesis of Campylobacter infection has lagged far behind that of other enteric pathogens. However, recent completion of the genome sequence of Campylobacter jejuni promises to open up the Campylobacter research field with the prospect of developing novel therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Ellen Crushell
- FRCPI, Children's Research Centre, Our Lady's Hospital for Sick Children, Crumlin, Dublin 12, Ireland.
| | | | | | | |
Collapse
|
43
|
Biswas D, Itoh K, Sasakawa C. Role of microfilaments and microtubules in the invasion of INT-407 cells by Campylobacter jejuni. Microbiol Immunol 2003; 47:469-73. [PMID: 12906108 DOI: 10.1111/j.1348-0421.2003.tb03372.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The internalization mechanisms triggered by Campylobacter jejuni were studied by invasion assays conducted with different inhibitors that act on the cytoskeleton structure of eukaryotic cells. The depolymerization of microfilaments by cytochalasin-D and that of microtubules by colchicines and nocodazole inhibited the uptake of C. jejuni into INT-407 cells in a dose-dependent manner. The inhibitory effect of microfilament depolymerization on C. jejuni internalization was more pronounced than that of microtubule depolymerization. By immunofluorescence microscopic observations, it was demonstrated that both microfilaments and microtubules were localized in INT-407 cells after C. jejuni infection. These data suggest that the internalization mechanism triggered by C. jejuni is associated with the combined effect of microfilaments and microtubules of host cells.
Collapse
Affiliation(s)
- Debabrata Biswas
- Laboratory of Veterinary Public Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | |
Collapse
|
44
|
Abstract
Campylobacter fetus is a Gram-negative bacterial pathogen of humans and ungulates and is normally transmitted via ingestion of contaminated food or water with infection resulting in mild to severe enteritis. However, despite clinical evidence that C. fetus infection often involves transient bacteremic states from which systemic infection may develop and the frequent isolation of C. fetus from extra-intestinal sites, this organism displays very poor invasiveness in in vitro models of infection. In this study, immunofluorescence microscopy and gentamicin protection assays were used to investigate the ability of six clinical isolates and one reference strain of C. fetus to adhere to and invade the human intestinal epithelial cell line, INT 407. During an initial 4-h infection period, all C. fetus strains were detected intracellularly using both techniques, though adherence and internalization levels were very low when determined from gentamicin protection assays. Microscopy results indicated that during a 4-h infection period, four of the five clinical strains tested were adherent to 41.3-87.3% of INT 407 cells observed and that 25.2-34.6% of INT 407 cells contained intracellular C. fetus. The C. fetus reference strain displayed the lowest levels of adherence and internalization. A modified infection assay revealed that C. fetus adherence did not necessarily culminate in internalization. Despite the large percentage of INT 407 cells with adherent bacteria, the percentage of INT 407 cells with intracellular bacteria remained unchanged when incubation was extended from 4 h to 20 h. However, microscopy of INT 407 cells 24 h postinfection (p.i.) revealed that infected host cells contained clusters of densely packed C. fetus cells. Gentamicin protection assays revealed that intracellular C. fetus cells were not only viable 24 h p.i. but also that C. fetus had increased in number approximately three- to fourfold between 4 and 24 h p.i., indicative of intracellular replication. Investigation of the role of the host cell cytoskeleton revealed that pretreatment of host cells with cytochalasin D, colchicine, vinblastine, taxol, or dimethyl sulfoxide (DMSO) did not impact upon C. fetus adherence or internalization of INT 407 cells. Microscopy indicated neither rearrangement nor colocalization of either microtubules or microfilaments in INT 407 cells in response to C. fetus adherence or internalization. Together, these data indicate that clinical isolates of C. fetus are capable of adhering, entering, and surviving within the nonphagocytic epithelial cell line, INT 407.
Collapse
Affiliation(s)
- Lori L Graham
- Department of Biology, St. Francis Xavier University, Antigonish, NS B2G 2W5, Canada.
| |
Collapse
|
45
|
Wasylnka JA, Moore MM. Uptake of Aspergillus fumigatus Conidia by phagocytic and nonphagocytic cells in vitro: quantitation using strains expressing green fluorescent protein. Infect Immun 2002; 70:3156-63. [PMID: 12011010 PMCID: PMC127978 DOI: 10.1128/iai.70.6.3156-3163.2002] [Citation(s) in RCA: 141] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Several pathogenic fungal organisms enter eukaryotic cells and manipulate the host cell environment to favor their own growth and survival. Aspergillus fumigatus is a saprophytic fungus that causes invasive lung disease in the immunocompromised host. To determine whether A. fumigatus could enter eukaryotic cells, we studied the uptake of two different GFP-expressing A. fumigatus strains into A549 lung epithelial cells, human umbilical vein endothelial (HUVE) cells, and J774 murine macrophages in vitro. A549 cells internalized 30% of the bound conidia whereas HUVE and J774 cells internalized 50 and 90%, respectively. Conidia within A549 cells remained viable for 6 h; however, 60 to 80% of conidia within J774 cells were killed after only 4 h. Live and heat-killed conidia were internalized to the same extent by A549 cells. After 6 h, almost none of the conidia inside A549 cells had germinated, whereas extracellular conidia had developed germ tubes. Internalization of conidia by A549 cells was a temperature-dependent process and required rearrangement of the underlying host cell cytoskeleton; uptake was inhibited by 75% with 0.5 microM cytochalasin D and by 65% with 5 microM colchicine. Fluorescent labeling of infected A549 cells with rhodamine phalloidin provided visible evidence of cytoskeletal alteration as many of the intracellular conidia were contained in actin-coated phagosomes. These data provide evidence that significant numbers of A. fumigatus conidia can be internalized by nonprofessional phagocytes in vitro and these cells may serve as reservoirs for immune cell evasion and dissemination throughout the host.
Collapse
Affiliation(s)
- Julie A Wasylnka
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada, V5A 1S6
| | | |
Collapse
|
46
|
Abstract
The important contribution of Campylobacter infections to human enteric disease is well established. Recent completion of the genomic sequence of a Campylobacter jejuni strain has heralded a renaissance in the field of Campylobacter pathogenesis research. With the application of novel, powerful technologies, our understanding of how these organisms mediate disease is set to evolve rapidly from its current, relatively neglected status.
Collapse
Affiliation(s)
- Billy Bourke
- Children's Research Center, Our Lady's Hospital for Sick Children, Department of Pediatrics, Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
47
|
Abstract
Campylobacter jejuni is the leading bacterial cause of food-borne illness worldwide and a major cause of Guillain-Barré paralysis. Recent molecular and cellular studies of one well-characterized C. jejuni strain have begun to unravel the details of an unusual microtubule-dependent (actin-filament-independent) gut-invasion mechanism, through which at least some C. jejuni initiate disease. Although responsible for causing a human dysenteric syndrome remarkably similar to that triggered by Shigella spp., current evidence suggests that C. jejuni use some markedly different molecular mechanisms of pathogenesis compared with shigellae.
Collapse
Affiliation(s)
- D J Kopecko
- Laboratory of Enteric and Sexually Transmitted Diseases, FDA-Center for Biologics Evaluation and Research, Bldg 29/420, NIH Campus, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|