1
|
Baztarrika I, Wösten MMSM, Alonso R, Martínez-Ballesteros I, Martinez-Malaxetxebarria I. Genes involved in the adhesion and invasion of Arcobacter butzleri. Microb Pathog 2024; 193:106752. [PMID: 38880315 DOI: 10.1016/j.micpath.2024.106752] [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: 12/17/2023] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Arcobacter butzleri is a foodborne pathogen that mainly causes enteritis in humans, but the number of cases of bacteraemia has increased in recent years. However, there is still limited knowledge on the pathogenic mechanisms of this bacterium. To investigate how A. butzleri causes disease, single knockout mutants were constructed in the cadF, ABU_RS00335, ciaB, and flaAB genes, which might be involved in adhesion and invasion properties. These mutants and the isogenic wild-type (WT) were then tested for their ability to adhere and invade human Caco-2 and HT29-MTX cells. The adhesion and invasion of A. butzleri RM4018 strain was also visualized by a Leica CTR 6500 confocal microscope. The adhesion and invasion abilities of mutants lacking the invasion antigen CiaB or a functional flagellum were lower than those of the WTs. However, the extent of the decrease varied depending on the strain and/or cell line. Mutants lacking the fibronectin (FN)-binding protein CadF consistently exhibited reduced abilities, while the inactivation of the other studied FN-binding protein, ABU_RS00335, led to a reduction in only one of the two strains tested. Therefore, the ciaB and flaAB genes appear to be important for A. butzleri adhesion and invasion properties, while cadF appears to be indispensable.
Collapse
Affiliation(s)
- Itsaso Baztarrika
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain
| | - Marc M S M Wösten
- Department Biomolecular Health Sciences, Utrecht University, Yalelaan 1, 3584, CL, Utrecht, the Netherlands
| | - Rodrigo Alonso
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain
| | - Ilargi Martínez-Ballesteros
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain
| | - Irati Martinez-Malaxetxebarria
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Álava, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents and Gene Therapy, 01006, Vitoria-Gasteiz, Álava, Spain.
| |
Collapse
|
2
|
Sharafutdinov I, Tegtmeyer N, Rohde M, Olofsson A, Rehman ZU, Arnqvist A, Backert S. Campylobacter jejuni Surface-Bound Protease HtrA, but Not the Secreted Protease nor Protease in Shed Membrane Vesicles, Disrupts Epithelial Cell-to-Cell Junctions. Cells 2024; 13:224. [PMID: 38334616 PMCID: PMC10854787 DOI: 10.3390/cells13030224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/01/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Fundamental functions of the intestinal epithelium include the digestion of food, absorption of nutrients, and its ability to act as the first barrier against intruding microbes. Campylobacter jejuni is a major zoonotic pathogen accounting for a substantial portion of bacterial foodborne illnesses. The germ colonizes the intestines of birds and is mainly transmitted to humans through the consumption of contaminated poultry meat. In the human gastrointestinal tract, the bacterium triggers campylobacteriosis that can progress to serious secondary disorders, including reactive arthritis, inflammatory bowel disease and Guillain-Barré syndrome. We recently discovered that C. jejuni serine protease HtrA disrupts intestinal epithelial barrier functions via cleavage of the tight and adherens junction components occludin, claudin-8 and E-cadherin. However, it is unknown whether epithelial damage is mediated by the secreted soluble enzyme, by HtrA contained in shed outer-membrane vesicles (OMVs) or by another mechanism that has yet to be identified. In the present study, we investigated whether soluble recombinant HtrA and/or purified OMVs induce junctional damage to polarized intestinal epithelial cells compared to live C. jejuni bacteria. By using electron and confocal immunofluorescence microscopy, we show that HtrA-expressing C. jejuni bacteria trigger efficient junctional cell damage, but not soluble purified HtrA or HtrA-containing OMVs, not even at high concentrations far exceeding physiological levels. Instead, we found that only bacteria with active protein biosynthesis effectively cleave junctional proteins, which is followed by paracellular transmigration of C. jejuni through the epithelial cell layer. These findings shed new light on the pathogenic activities of HtrA and virulence strategies of C. jejuni.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058 Erlangen, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058 Erlangen, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Inhoffenstraße 7, D-38124 Braunschweig, Germany
| | - Annelie Olofsson
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
| | - Zia ur Rehman
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
| | - Anna Arnqvist
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058 Erlangen, Germany
| |
Collapse
|
3
|
Teixeira SC, Teixeira TL, Tavares PCB, Alves RN, da Silva AA, Borges BC, Martins FA, Dos Santos MA, de Castilhos P, E Silva Brígido RT, Notário AFO, Silveira ACA, da Silva CV. Subversion strategies of lysosomal killing by intracellular pathogens. Microbiol Res 2023; 277:127503. [PMID: 37748260 DOI: 10.1016/j.micres.2023.127503] [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: 07/16/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 09/27/2023]
Abstract
Many pathogenic organisms need to reach either an intracellular compartment or the cytoplasm of a target cell for their survival, replication or immune system evasion. Intracellular pathogens frequently penetrate into the cell through the endocytic and phagocytic pathways (clathrin-mediated endocytosis, phagocytosis and macropinocytosis) that culminates in fusion with lysosomes. However, several mechanisms are triggered by pathogenic microorganisms - protozoan, bacteria, virus and fungus - to avoid destruction by lysosome fusion, such as rupture of the phagosome and thereby release into the cytoplasm, avoidance of autophagy, delaying in both phagolysosome biogenesis and phagosomal maturation and survival/replication inside the phagolysosome. Here we reviewed the main data dealing with phagosome maturation and evasion from lysosomal killing by different bacteria, protozoa, fungi and virus.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Thaise Lara Teixeira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | - Aline Alves da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Bruna Cristina Borges
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Flávia Alves Martins
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marlus Alves Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Patrícia de Castilhos
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | | | | | | | - Claudio Vieira da Silva
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil.
| |
Collapse
|
4
|
Molecular Targets in Campylobacter Infections. Biomolecules 2023; 13:biom13030409. [PMID: 36979344 PMCID: PMC10046527 DOI: 10.3390/biom13030409] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Human campylobacteriosis results from foodborne infections with Campylobacter bacteria such as Campylobacter jejuni and Campylobacter coli, and represents a leading cause of bacterial gastroenteritis worldwide. After consumption of contaminated poultry meat, constituting the major source of pathogenic transfer to humans, infected patients develop abdominal pain and diarrhea. Post-infectious disorders following acute enteritis may occur and affect the nervous system, the joints or the intestines. Immunocompromising comorbidities in infected patients favor bacteremia, leading to vascular inflammation and septicemia. Prevention of human infection is achieved by hygiene measures focusing on the reduction of pathogenic food contamination. Molecular targets for the treatment and prevention of campylobacteriosis include bacterial pathogenicity and virulence factors involved in motility, adhesion, invasion, oxygen detoxification, acid resistance and biofilm formation. This repertoire of intervention measures has recently been completed by drugs dampening the pro-inflammatory immune responses induced by the Campylobacter endotoxin lipo-oligosaccharide. Novel pharmaceutical strategies will combine anti-pathogenic and anti-inflammatory effects to reduce the risk of both anti-microbial resistance and post-infectious sequelae of acute enteritis. Novel strategies and actual trends in the combat of Campylobacter infections are presented in this review, alongside molecular targets applied for prevention and treatment strategies.
Collapse
|
5
|
Campylobacter jejuni Modulates Reactive Oxygen Species Production and NADPH Oxidase 1 Expression in Human Intestinal Epithelial Cells. Cell Microbiol 2023. [DOI: 10.1155/2023/3286330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Campylobacter jejuni is the major bacterial cause of foodborne gastroenteritis worldwide. Mechanistically, how this pathogen interacts with intrinsic defence machinery of human intestinal epithelial cells (IECs) remains elusive. To address this, we investigated how C. jejuni counteracts the intracellular and extracellular reactive oxygen species (ROS) in IECs. Our work shows that C. jejuni differentially regulates intracellular and extracellular ROS production in human T84 and Caco-2 cells. C. jejuni downregulates the transcription and translation of nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase (NOX1), a key ROS-generating enzyme in IECs and antioxidant defence genes CAT and SOD1. Furthermore, inhibition of NOX1 by diphenylene iodonium (DPI) and siRNA reduced C. jejuni ability to interact, invade, and intracellularly survive within T84 and Caco-2 cells. Collectively, these findings provide mechanistic insight into how C. jejuni modulates the IEC defence machinery.
Collapse
|
6
|
Prevalence, drug resistance spectrum and virulence gene analysis of Campylobacter jejuni in broiler farms in central Shanxi, China. Poult Sci 2022; 102:102419. [PMID: 36599220 PMCID: PMC9823222 DOI: 10.1016/j.psj.2022.102419] [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] [Received: 10/28/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
This study collected 324 chicken cloacal swabs from 6 broiler farms in 4 different areas in Shanxi Province, China (i.e., Lvliang, Taiyuan, Jinzhong, and Yangquan), and analyzed the antimicrobial resistance and virulence-associated genes of the isolates to investigate the prevalence, drug resistance, and virulence gene data of Campylobacter jejuni in broilers. The population structure of C. jejuni and genetic evolutionary relationships among isolates from broiler farms in different regions were studied by using multilocus sequence typing. A total of 35 C. jejuni isolates with an infection rate of 10.8% (35/324) were obtained. The isolates were most resistant to ampicillin (85.7%) and were most sensitive to erythromycin (14.3%). Isolates with multidrug resistance accounted for 88.6% of the total isolates. In this experiment, 15 distinct sequence types were identified and included 9 new unique sequence types. cadF was present in all isolates, and ciaB had the lowest prevalence (51.4%). C. jejuni collected from broiler farms in central Shanxi had varied infection rates, and their overall positive rate was lower than of C. jejuni collected from other regions of the country. The isolates had high resistance to quinolones and β-lactams, and multidrug resistance was prevalent. The isolates were genotypically diverse and carried 5 virulence-associated genes at high rates. Therefore, the importance of source contamination control in broiler farms is emphasized and may have considerable effects on human and animal health.
Collapse
|
7
|
Role of Host Small GTPases in Apicomplexan Parasite Infection. Microorganisms 2022; 10:microorganisms10071370. [PMID: 35889089 PMCID: PMC9319929 DOI: 10.3390/microorganisms10071370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
The Apicomplexa are obligate intracellular parasites responsible for several important human diseases. These protozoan organisms have evolved several strategies to modify the host cell environment to create a favorable niche for their survival. The host cytoskeleton is widely manipulated during all phases of apicomplexan intracellular infection. Moreover, the localization and organization of host organelles are altered in order to scavenge nutrients from the host. Small GTPases are a class of proteins widely involved in intracellular pathways governing different processes, from cytoskeletal and organelle organization to gene transcription and intracellular trafficking. These proteins are already known to be involved in infection by several intracellular pathogens, including viruses, bacteria and protozoan parasites. In this review, we recapitulate the mechanisms by which apicomplexan parasites manipulate the host cell during infection, focusing on the role of host small GTPases. We also discuss the possibility of considering small GTPases as potential targets for the development of novel host-targeted therapies against apicomplexan infections.
Collapse
|
8
|
Javed K, Gul F, Abbasi R, Batool S, Noreen Z, Bokhari H, Javed S. In Silico and In Vitro Analysis of Helicobacter pullorum Type Six Secretory Protein Hcp and Its Role in Bacterial Invasion and Pathogenesis. Curr Microbiol 2022; 79:195. [PMID: 35593885 DOI: 10.1007/s00284-022-02892-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/26/2022] [Indexed: 11/28/2022]
Abstract
Helicobacter pullorum is a human zoonotic pathogen transmitted through poultry where it is associated with vibrionic hepatitis and colitis. Hemolysin co-regulated protein (Hcp) is an important structural as well as effector protein of type six secretory system; however, its role in H. pullorum invasion and pathogenesis has not been elucidated. In this study, we predicted the Helicobacter pullorum Hcp (HpuHcp) structure and identified Campylobacter jejuni Hcp (CjHcp) as its nearest homologue. Analysis of the predicted structure shows several common bacterial Hcp motifs like Protein kinase C phosphorylation site, Casein kinase II phosphorylation site, N-myristoylation site, cAMP-and cCGMP-dependent protein kinase phosphorylation site, N-glycosylation site. The presence of unique microbodies C-terminal targeting signal domain was present in HpuHcp which was seen for the first time in CjHcp. This could indicate that Hcp is a structural protein as well as a secretory protein. Moreover, the presence of a deamidase domain, similar to the tecA of Burkholderia cenocepacia an opportunistic pathogen, may help in bacterial internalization as it depolymerises the membranous actin by deamidation of the host cell Rho GTPases cdc42 and Rac1, which was supported by increased invasion of hepatocytes by Hcp-positive isolates.
Collapse
Affiliation(s)
- Kashaf Javed
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan
| | - Farzana Gul
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan
| | - Rashda Abbasi
- Institute of Biomedical and Genetic Engineering, Islamabad, Pakistan
| | - Sidra Batool
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan.,Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Zobia Noreen
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan
| | - Habib Bokhari
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan.,Bioscience Department, Kohsar University Murree, Near Kashmir Point, Murree, Punjab, Pakistan
| | - Sundus Javed
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan.
| |
Collapse
|
9
|
Campylobacter jejuni Serine Protease HtrA Induces Paracellular Transmigration of Microbiota across Polarized Intestinal Epithelial Cells. Biomolecules 2022; 12:biom12040521. [DOI: 10.3390/biom12040521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 02/05/2023] Open
Abstract
Campylobacter jejuni represents an eminent zoonotic germ responsible for foodborne infections causing campylobacteriosis. In addition, infections with C. jejuni constitute a risk factor for the occurrence of inflammatory bowel disease (IBD). In the latter case, patients show inflammatory reactions not only against C. jejuni, but also against the non-infectious microbiota. However, the involved mechanisms and molecular basis are still largely unclear. We recently reported that C. jejuni breaches the intestinal epithelial barrier by secretion of serine protease HtrA (high temperature requirement A), which cleaves several major tight and adherens junction proteins. In the present study, we aimed to study if HtrA-expressing C. jejuni may also trigger the transepithelial migration of non-pathogenic gastrointestinal microbiota. Using confocal immunofluorescence and scanning electron microscopy, we demonstrate that C. jejuni wild-type (wt) as well as the isogenic ∆htrA mutant bind to the surface of polarized intestinal Caco-2 epithelial cells, but do not invade them at the apical side. Instead, C. jejuni wt, but not ∆htrA mutant, disrupt the cellular junctions and transmigrate using the paracellular route between neighboring cells. Using transwell filter systems, we then co-incubated the cells with C. jejuni and non-invasive microbiota strains, either Escherichia coli or Lactococcus lactis. Interestingly, C. jejuni wt, but not ∆htrA mutant, induced the efficient transmigration of these microbiota bacteria into the basal compartment. Thus, infection of the intestinal epithelium with C. jejuni causes local opening of cellular junctions and paracellular translocation in an HtrA-dependent manner, which paves the way for transmigration of microbiota that is otherwise non-invasive. Taken together, these findings may have impacts on various Campylobacter-associated diseases such as IBD, which are discussed here.
Collapse
|
10
|
Fukushima S, Shimohata T, Inoue Y, Kido J, Uebanso T, Mawatari K, Takahashi A. Recruitment of LC3 by Campylobacter jejuni to Bacterial Invasion Site on Host Cells via the Rac1-Mediated Signaling Pathway. Front Cell Infect Microbiol 2022; 12:829682. [PMID: 35310852 PMCID: PMC8927770 DOI: 10.3389/fcimb.2022.829682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/02/2022] [Indexed: 11/29/2022] Open
Abstract
Campylobacter jejuni is a leading cause of food-borne disease worldwide. The pathogenicity of C. jejuni is closely associated with the internalization process in host epithelial cells, which is related to a host immune response. Autophagy indicates a key role in the innate immune system of the host to exclude invasive pathogens. Most bacteria are captured by autophagosomes and degraded by autophagosome-lysosome fusion in host cells. However, several pathogens, such as Salmonella and Shigella, avoid and/or escape autophagic degradation to establish infection. But autophagy involvement as a host immune response to C. jejuni infection has not been clarified. This study revealed autophagy association in C. jejuni infection. During infection, C. jejuni activated the Rho family small GTPase Rac1 signaling pathway, which modulates actin remodeling and promotes the internalization of this pathogen. In this study, we found the LC3 contribution to C. jejuni invasion signaling via the Rac1 signaling pathway. Interestingly, during C. jejuni invasion, LC3 was recruited to bacterial entry site depending on Rac1 GTPase activation just at the early step of the infection. C. jejuni infection induced LC3-II conversion, and autophagy induction facilitated C. jejuni internalization. Also, autophagy inhibition attenuated C. jejuni invasion step. Moreover, Rac1 recruited LC3 to the cellular membrane, activating the invasion of C. jejuni. Altogether, our findings provide insights into the new function of LC3 in bacterial invasion. We found the interaction between the Rho family small GTPase, Rac1, and autophagy-associated protein, LC3.
Collapse
Affiliation(s)
- Shiho Fukushima
- Department of Preventive Environment and Nutrition Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takaaki Shimohata
- Department of Preventive Environment and Nutrition Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
- Faculty of Marine Biosciences, Fukui Prefectural University, Fukui, Japan
- *Correspondence: Takaaki Shimohata, ;
| | - Yuri Inoue
- Department of Preventive Environment and Nutrition Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Junko Kido
- Department of Preventive Environment and Nutrition Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takashi Uebanso
- Department of Preventive Environment and Nutrition Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Akira Takahashi
- Department of Preventive Environment and Nutrition Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
11
|
Cox CA, Bogacz M, El Abbar FM, Browning DD, Hsueh BY, Waters CM, Lee VT, Thompson SA. The Campylobacter jejuni Response Regulator and Cyclic-Di-GMP Binding CbrR Is a Novel Regulator of Flagellar Motility. Microorganisms 2021; 10:microorganisms10010086. [PMID: 35056537 PMCID: PMC8779298 DOI: 10.3390/microorganisms10010086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/19/2021] [Accepted: 12/29/2021] [Indexed: 01/03/2023] Open
Abstract
A leading cause of bacterial gastroenteritis, Campylobacter jejuni is also associated with broad sequelae, including extragastrointestinal conditions such as reactive arthritis and Guillain-Barré Syndrome (GBS). CbrR is a C. jejuni response regulator that is annotated as a diguanylate cyclase (DGC), an enzyme that catalyzes the synthesis of c-di-GMP, a universal bacterial second messenger, from GTP. In C. jejuni DRH212, we constructed an unmarked deletion mutant, cbrR-, and complemented mutant, cbrR+. Motility assays indicated a hyper-motile phenotype associated with cbrR-, whereas motility was deficient in cbrR+. The overexpression of CbrR in cbrR+ was accompanied by a reduction in expression of FlaA, the major flagellin. Biofilm assays and scanning electron microscopy demonstrated similarities between DRH212 and cbrR-; however, cbrR+ was unable to form significant biofilms. Transmission electron microscopy showed similar cell morphology between the three strains; however, cbrR+ cells lacked flagella. Differential radial capillary action of ligand assays (DRaCALA) showed that CbrR binds GTP and c-di-GMP. Liquid chromatography tandem mass spectrometry detected low levels of c-di-GMP in C. jejuni and in E. coli expressing CbrR. CbrR is therefore a negative regulator of FlaA expression and motility, a critical virulence factor in C. jejuni pathogenesis.
Collapse
Affiliation(s)
- Claudia A. Cox
- Department of Medicine, Division of Infectious Diseases, Augusta University, Augusta, GA 30912, USA; (C.A.C.); (M.B.); (F.M.E.A.)
| | - Marek Bogacz
- Department of Medicine, Division of Infectious Diseases, Augusta University, Augusta, GA 30912, USA; (C.A.C.); (M.B.); (F.M.E.A.)
| | - Faiha M. El Abbar
- Department of Medicine, Division of Infectious Diseases, Augusta University, Augusta, GA 30912, USA; (C.A.C.); (M.B.); (F.M.E.A.)
| | - Darren D. Browning
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA;
| | - Brian Y. Hsueh
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA; (B.Y.H.); (C.M.W.)
| | - Chris M. Waters
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA; (B.Y.H.); (C.M.W.)
| | - Vincent T. Lee
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| | - Stuart A. Thompson
- Department of Medicine, Division of Infectious Diseases, Augusta University, Augusta, GA 30912, USA; (C.A.C.); (M.B.); (F.M.E.A.)
- Correspondence:
| |
Collapse
|
12
|
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
|
13
|
Negretti NM, Gourley CR, Talukdar PK, Clair G, Klappenbach CM, Lauritsen CJ, Adkins JN, Konkel ME. The Campylobacter jejuni CiaD effector co-opts the host cell protein IQGAP1 to promote cell entry. Nat Commun 2021; 12:1339. [PMID: 33637714 PMCID: PMC7910587 DOI: 10.1038/s41467-021-21579-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Campylobacter jejuni is a foodborne pathogen that binds to and invades the epithelial cells lining the human intestinal tract. Maximal invasion of host cells by C. jejuni requires cell binding as well as delivery of the Cia proteins (Campylobacter invasion antigens) to the host cell cytosol via the flagellum. Here, we show that CiaD binds to the host cell protein IQGAP1 (a Ras GTPase-activating-like protein), thus displacing RacGAP1 from the IQGAP1 complex. This, in turn, leads to the unconstrained activity of the small GTPase Rac1, which is known to have roles in actin reorganization and internalization of C. jejuni. Our results represent the identification of a host cell protein targeted by a flagellar secreted effector protein and demonstrate that C. jejuni-stimulated Rac signaling is dependent on IQGAP1.
Collapse
Affiliation(s)
- Nicholas M Negretti
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Christopher R Gourley
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Prabhat K Talukdar
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Geremy Clair
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Courtney M Klappenbach
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Cody J Lauritsen
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Joshua N Adkins
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Michael E Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| |
Collapse
|
14
|
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
|
15
|
den Hartog G, Butcher LD, Ablack AL, Pace LA, Ablack JNG, Xiong R, Das S, Stappenbeck TS, Eckmann L, Ernst PB, Crowe SE. Apurinic/Apyrimidinic Endonuclease 1 Restricts the Internalization of Bacteria Into Human Intestinal Epithelial Cells Through the Inhibition of Rac1. Front Immunol 2021; 11:553994. [PMID: 33603730 PMCID: PMC7884313 DOI: 10.3389/fimmu.2020.553994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Pathogenic intestinal bacteria lead to significant disease in humans. Here we investigated the role of the multifunctional protein, Apurinic/apyrimidinic endonuclease 1 (APE1), in regulating the internalization of bacteria into the intestinal epithelium. Intestinal tumor-cell lines and primary human epithelial cells were infected with Salmonella enterica serovar Typhimurium or adherent-invasive Escherichia coli. The effects of APE1 inhibition on bacterial internalization, the regulation of Rho GTPase Rac1 as well as the epithelial cell barrier function were assessed. Increased numbers of bacteria were present in APE1-deficient colonic tumor cell lines and primary epithelial cells. Activation of Rac1 was augmented following infection but negatively regulated by APE1. Pharmacological inhibition of Rac1 reversed the increase in intracellular bacteria in APE1-deficient cells whereas overexpression of constitutively active Rac1 augmented the numbers in APE1-competent cells. Enhanced numbers of intracellular bacteria resulted in the loss of barrier function and a delay in its recovery. Our data demonstrate that APE1 inhibits the internalization of invasive bacteria into human intestinal epithelial cells through its ability to negatively regulate Rac1. This activity also protects epithelial cell barrier function.
Collapse
Affiliation(s)
- Gerco den Hartog
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Lindsay D Butcher
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Amber L Ablack
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Laura A Pace
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Jailal N G Ablack
- Department of Medicine, Division of Rheumatology, University of California San Diego, La Jolla, CA, United States
| | - Richard Xiong
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Soumita Das
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States
| | | | - Lars Eckmann
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States
| | - Peter B Ernst
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Center for Mucosal Immunology, Allergy and Vaccine Development, Department of Pathology, University of California San Diego, La Jolla, CA, United States.,Department of Immunology, Chiba University, Chiba, Japan
| | - Sheila E Crowe
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA, United States.,Division of ImmunoBiology, Washington University, St. Louis, MO, United States
| |
Collapse
|
16
|
Modeling Invasion of Campylobacter jejuni into Human Small Intestinal Epithelial-Like Cells by Bayesian Inference. Appl Environ Microbiol 2020; 87:AEM.01551-20. [PMID: 33067190 DOI: 10.1128/aem.01551-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023] Open
Abstract
Current approaches used for dose-response modeling of low-dose exposures of pathogens rely on assumptions and extrapolations. These models are important for quantitative microbial risk assessment of food. A mechanistic framework has been advocated as an alternative approach for evaluating dose-response relationships. The objectives of this study were to investigate the invasion behavior of Campylobacter jejuni, which could arise as a foodborne illness even if there are low counts of pathogens, into Caco-2 cells as a model of intestinal cells and to develop a mathematical model for invading cell counts to reveal a part of the infection dose-response mechanism. Monolayer-cultured Caco-2 cells and various concentrations of C. jejuni in culture were cocultured for up to 12 h. The numbers of C. jejuni bacteria invading Caco-2 cells were determined after coculture for different time periods. There appeared to be a maximum limit to the invading bacterial counts, which showed an asymptotic exponential increase. The invading bacterial counts were higher with higher exposure concentrations (maximum, 5.0 log CFU/cm2) than with lower exposure concentrations (minimum, 0.6 log CFU/cm2). In contrast, the ratio of invading bacteria (number of invading bacteria divided by the total number of bacteria exposed) showed a similar trend regardless of the exposure concentration. Invasion of C. jejuni into intestinal cells was successfully demonstrated and described by the developed differential equation model with Bayesian inference. The model accuracy showed that the 99% prediction band covered more than 97% of the observed values. These findings provide important information on mechanistic pathogen dose-response relationships and an alternative approach for dose-response modeling.IMPORTANCE One of the infection processes of C. jejuni, the invasion behavior of the bacteria in intestinal epithelial cells, was revealed, and a mathematical model for prediction of the cell-invading pathogen counts was developed for the purpose of providing part of a dose-response model for C. jejuni based on the infection mechanism. The developed predictive model showed a high accuracy of more than 97% and successfully described the C. jejuni invading counts. The bacterial invasion predictive model of this study will be essential for the development of a dose-response model for C. jejuni based on the infection mechanism.
Collapse
|
17
|
Sharafutdinov I, Esmaeili DS, Harrer A, Tegtmeyer N, Sticht H, Backert S. Campylobacter jejuni Serine Protease HtrA Cleaves the Tight Junction Component Claudin-8. Front Cell Infect Microbiol 2020; 10:590186. [PMID: 33364202 PMCID: PMC7752809 DOI: 10.3389/fcimb.2020.590186] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Campylobacter jejuni express the high temperature requirement protein A (HtrA), a secreted serine protease, which is implicated in virulence properties of the pathogen. Previous studies have shown that C. jejuni HtrA can cleave the epithelial transmembrane proteins occludin and E-cadherin in the tight and adherens junctions, respectively. In the present report, we studied the interaction of HtrA with another human tight junction protein, claudin-8. Confocal immunofluorescence experiments have shown that C. jejuni infection of the intestinal polarized epithelial cells in vitro leads to a relocation of claudin-8. Wild-type C. jejuni induced the downregulation of claudin-8 signals in the tight junctions and an accumulation of claudin-8 agglomerates in the cytoplasm, which were not seen during infection with isogenic ΔhtrA knockout deletion or protease-inactive S197A point mutants. Western blotting of protein samples from infected vs. uninfected cells revealed that an 18-kDa carboxy-terminal fragment is cleaved-off from the 26-kDa full-length claudin-8 protein, but not during infection with the isogenic ΔhtrA mutant. These results were confirmed by in vitro cleavage assays using the purified recombinant C. jejuni HtrA and human claudin-8 proteins. Recombinant HtrA cleaved purified claudin-8 in vitro giving rise to the same 18-kDa sized carboxy-terminal cleavage product. Mapping studies revealed that HtrA cleavage occurs in the first extracellular loop of claudin-8. Three-dimensional modeling of the claudin-8 structure identified an exposed HtrA cleavage site between the amino acids alanine 58 and asparagine 59, which is in well agreement with the mapping studies. Taken together, HtrA operates as a secreted virulence factor targeting multiple proteins both in the tight and adherens junctions. This strategy may help the bacteria to open the cell-to-cell junctions, and to transmigrate across the intestinal epithelium by a paracellular mechanism and establish an acute infection.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Delara Soltan Esmaeili
- Department of Biology, Division of Microbiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Aileen Harrer
- Department of Biology, Division of Microbiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
18
|
Kreling V, Falcone FH, Kehrenberg C, Hensel A. Campylobacter sp.: Pathogenicity factors and prevention methods-new molecular targets for innovative antivirulence drugs? Appl Microbiol Biotechnol 2020; 104:10409-10436. [PMID: 33185702 PMCID: PMC7662028 DOI: 10.1007/s00253-020-10974-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/24/2020] [Accepted: 10/21/2020] [Indexed: 02/08/2023]
Abstract
Infections caused by bacterial species from the genus Campylobacter are one of the four main causes of strong diarrheal enteritis worldwide. Campylobacteriosis, a typical food-borne disease, can range from mild symptoms to fatal illness. About 550 million people worldwide suffer from campylobacteriosis and lethality is about 33 million p.a. This review summarizes the state of the current knowledge on Campylobacter with focus on its specific virulence factors. Using this knowledge, multifactorial prevention strategies can be implemented to reduce the prevalence of Campylobacter in the food chain. In particular, antiadhesive strategies with specific adhesion inhibitors seem to be a promising concept for reducing Campylobacter bacterial load in poultry production. Antivirulence compounds against bacterial adhesion to and/or invasion into the host cells can open new fields for innovative antibacterial agents. Influencing chemotaxis, biofilm formation, quorum sensing, secretion systems, or toxins by specific inhibitors can help to reduce virulence of the bacterium. In addition, the unusual glycosylation of the bacterium, being a prerequisite for effective phase variation and adaption to different hosts, is yet an unexplored target for combating Campylobacter sp. Plant extracts are widely used remedies in developing countries to combat infections with Campylobacter. Therefore, the present review summarizes the use of natural products against the bacterium in an attempt to stimulate innovative research concepts on the manifold still open questions behind Campylobacter towards improved treatment and sanitation of animal vectors, treatment of infected patients, and new strategies for prevention. KEY POINTS: • Campylobacter sp. is a main cause of strong enteritis worldwide. • Main virulence factors: cytolethal distending toxin, adhesion proteins, invasion machinery. • Strong need for development of antivirulence compounds.
Collapse
Affiliation(s)
- Vanessa Kreling
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Franco H Falcone
- Institute of Parasitology, University of Gießen, Schubertstraße 81, 35392, Gießen, Germany
| | - Corinna Kehrenberg
- Institute of Veterinary Food Science, University of Gießen, Frankfurterstraße 81, 35392, Gießen, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany.
| |
Collapse
|
19
|
Negretti NM, Ye Y, Malavasi LM, Pokharel SM, Huynh S, Noh S, Klima CL, Gourley CR, Ragle CA, Bose S, Looft T, Parker CT, Clair G, Adkins JN, Konkel ME. A porcine ligated loop model reveals new insight into the host immune response against Campylobacter jejuni. Gut Microbes 2020; 12:1-25. [PMID: 32887530 PMCID: PMC7524355 DOI: 10.1080/19490976.2020.1814121] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 02/03/2023] Open
Abstract
The symptoms of infectious diarrheal disease are mediated by a combination of a pathogen's virulence factors and the host immune system. Campylobacter jejuni is the leading bacterial cause of diarrhea worldwide due to its near-ubiquitous zoonotic association with poultry. One of the outstanding questions is to what extent the bacteria are responsible for the diarrheal symptoms via intestinal cell necrosis versus immune cell initiated tissue damage. To determine the stepwise process of inflammation that leads to diarrhea, we used a piglet ligated intestinal loop model to study the intestinal response to C. jejuni. Pigs were chosen due to the anatomical similarity between the porcine and the human intestine. We found that the abundance of neutrophil related proteins increased in the intestinal lumen during C. jejuni infection, including proteins related to neutrophil migration (neutrophil elastase and MMP9), actin reorganization (Arp2/3), and antimicrobial proteins (lipocalin-2, myeloperoxidase, S100A8, and S100A9). The appearance of neutrophil proteins also corresponded with increases of the inflammatory cytokines IL-8 and TNF-α. Compared to infection with the C. jejuni wild-type strain, infection with the noninvasive C. jejuni ∆ciaD mutant resulted in a blunted inflammatory response, with less inflammatory cytokines and neutrophil markers. These findings indicate that intestinal inflammation is driven by C. jejuni virulence and that neutrophils are the predominant cell type responding to C. jejuni infection. We propose that this model can be used as a platform to study the early immune events during infection with intestinal pathogens.
Collapse
Affiliation(s)
- Nicholas M Negretti
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Yinyin Ye
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lais M Malavasi
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Swechha M Pokharel
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Steven Huynh
- Produce Safety and Microbiology, United States Department of Agriculture-Agricultural Research Service, Albany, CA, USA
| | - Susan Noh
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, USA
- Washington Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Cassidy L Klima
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, USA
| | - Christopher R Gourley
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Claude A Ragle
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Santanu Bose
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Torey Looft
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, USA
| | - Craig T Parker
- Produce Safety and Microbiology, United States Department of Agriculture-Agricultural Research Service, Albany, CA, USA
| | - Geremy Clair
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joshua N Adkins
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Michael E Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
20
|
Yang WJ, Yan JB, Zhang L, Zhao F, Mei ZM, Yang YN, Xiang Y, Xing YQ. Paxillin promotes the migration and angiogenesis of HUVECs and affects angiogenesis in the mouse cornea. Exp Ther Med 2020; 20:901-909. [PMID: 32742332 PMCID: PMC7388276 DOI: 10.3892/etm.2020.8751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/07/2019] [Indexed: 12/20/2022] Open
Abstract
Neonatal vascular ophthalmopathy is a refractory ophthalmologic disease, and is a major cause of blindness. Occurrence of neonatal vascular ophthalmopathy may be associated with Paxillin, a cellular adhesion molecule which promotes the migration of endothelial cells and angiogenesis. To explore the role of PXN in corneal angiogenesis, human umbilical vein endothelial cells were divided into five groups: i) Control group; ii) Empty vector-transfected control group; iii) PXN knockdown group (shPXN group); iv) PXN-negative control (NC) group; and v) PXN over-expressed group (overExp group). PXN protein levels, migration and tube formation were assessed in the different experimental groups. Mice were divided into four groups: i) Control; ii) Model; iii) shPXN; and iv) overExp groups. Tube formation was significantly increased in the overExp group compared with the empty vector-transfected control group (P<0.01). Tube formation was significantly decreased in the shPXN group compared with the PXN-NC group (P<0.01). In mice, blood corpuscles were significantly decreased in the shPXN group. PXN promoted the migration of endothelial cells and corneal angiogenesis. The results of the present study suggest a role for PXN in corneal angiogenesis and provide a theoretical basis and potential target for the treatment of corneal angiogenesis.
Collapse
Affiliation(s)
- Wan-Ju Yang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jiang-Bo Yan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Li Zhang
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Fang Zhao
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Zhong-Ming Mei
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yan-Ning Yang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yi Xiang
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yi-Qiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
21
|
Konkel ME, Talukdar PK, Negretti NM, Klappenbach CM. Taking Control: Campylobacter jejuni Binding to Fibronectin Sets the Stage for Cellular Adherence and Invasion. Front Microbiol 2020; 11:564. [PMID: 32328046 PMCID: PMC7161372 DOI: 10.3389/fmicb.2020.00564] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/16/2020] [Indexed: 12/18/2022] Open
Abstract
Campylobacter jejuni, a foodborne pathogen, is one of the most common bacterial causes of gastroenteritis in the world. Undercooked poultry, raw (unpasteurized) dairy products, untreated water, and contaminated produce are the most common sources associated with infection. C. jejuni establishes a niche in the gut by adhering to and invading epithelial cells, which results in diarrhea with blood and mucus in the stool. The process of colonization is mediated, in part, by surface-exposed molecules (adhesins) that bind directly to host cell ligands or the extracellular matrix (ECM) surrounding cells. In this review, we introduce the known and putative adhesins of the foodborne pathogen C. jejuni. We then focus our discussion on two C. jejuni Microbial Surface Components Recognizing Adhesive Matrix Molecule(s) (MSCRAMMs), termed CadF and FlpA, which have been demonstrated to contribute to C. jejuni colonization and pathogenesis. In vitro studies have determined that these two surface-exposed proteins bind to the ECM glycoprotein fibronectin (FN). In vivo studies have shown that cadF and flpA mutants exhibit impaired colonization of chickens compared to the wild-type strain. Additional studies have revealed that CadF and FlpA stimulate epithelial cell signaling pathways necessary for cell invasion. Interestingly, CadF and FlpA have distinct FN-binding domains, suggesting that the functions of these proteins are non-redundant. In summary, the binding of FN by C. jejuni CadF and FlpA adhesins has been demonstrated to contribute to adherence, invasion, and cell signaling.
Collapse
Affiliation(s)
- Michael E. Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | | | | | | |
Collapse
|
22
|
Talukdar PK, Negretti NM, Turner KL, Konkel ME. Molecular Dissection of the Campylobacter jejuni CadF and FlpA Virulence Proteins in Binding to Host Cell Fibronectin. Microorganisms 2020; 8:E389. [PMID: 32168837 PMCID: PMC7143056 DOI: 10.3390/microorganisms8030389] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023] Open
Abstract
Campylobacter jejuni, a zoonotic pathogen that frequently colonizes poultry, possesses two Microbial Surface Components Recognizing Adhesive Matrix Molecule(s) (MSCRAMMs) termed CadF and FlpA that bind to the glycoprotein fibronectin (FN). Previous to this study, it was not known whether the CadF and FlpA proteins were functionally redundant or if both were required to potentiate host cell binding and signaling processes. We addressed these questions by generating a complete repertoire of cadF and flpA mutants and complemented isolates, and performing multiple phenotypic assays. Both CadF and FlpA were found to be necessary for the maximal binding of C. jejuni to FN and to host cells. In addition, both CadF and FlpA are required for the delivery of the C. jejuni Cia effector proteins into the cytosol of host target cells, which in turn activates the MAPK signaling pathway (Erk 1/2) that is required for the C. jejuni invasion of host cells. These data demonstrate the non-redundant and bi-functional nature of these two C. jejuni FN-binding proteins. Taken together, the C. jejuni CadF and FlpA adhesins facilitate the binding of C. jejuni to the host cells, permit delivery of effector proteins into the cytosol of a host target cell, and aid in the rewiring of host cell signaling pathways to alter host cell behavior.
Collapse
Affiliation(s)
| | | | | | - Michael E. Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-7520, USA; (P.K.T.); (N.M.N.); (K.L.T.)
| |
Collapse
|
23
|
Butkevych E, Lobo de Sá FD, Nattramilarasu PK, Bücker R. Contribution of Epithelial Apoptosis and Subepithelial Immune Responses in Campylobacter jejuni- Induced Barrier Disruption. Front Microbiol 2020; 11:344. [PMID: 32210941 PMCID: PMC7067706 DOI: 10.3389/fmicb.2020.00344] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Campylobacter jejuni is a widespread zoonotic pathogen and the leading bacterial cause of foodborne gastroenteritis in humans. Previous infection studies showed disruption of intercellular contacts, induction of epithelial apoptosis, and immune activation, all three contributing to intestinal barrier dysfunction leading to diarrhea. The present study aims to determine the impact of subepithelial immune cells on intestinal barrier dysfunction during Campylobacter jejuni infection and the underlying pathological mechanisms. Infection was performed in a co-culture of confluent monolayers of the human colon cell line HT-29/B6-GR/MR and THP-1 immune cells. Twenty-two hours after infection, transepithelial electrical resistance (TER) was decreased by 58 ± 6% compared to controls. The infection resulted in an increase in permeability for fluorescein (332 Da; 4.5-fold) and for FITC-dextran (4 kDa; 3.5-fold), respectively. In contrast, incubation of the co-culture with the pan-caspase inhibitor Q-VD-OPh during the infection resulted in a complete recovery of the decrease in TER and a normalization of flux values. Fluorescence microscopy showed apoptotic fragmentation in infected cell monolayers resulting in a 5-fold increase of the apoptotic ratio, accompanied by an increased caspase-3 cleavage and caspase-3/7 activity, which both were not present after Q-VD-OPh treatment. Western blot analysis revealed increased claudin-1 and claudin-2 protein expression. Inhibition of apoptosis induction did not normalize these tight junction changes. TNFα concentration was increased during the infection in the co-culture. In conclusion, Campylobacter jejuni infection and the consequent subepithelial immune activation cause intestinal barrier dysfunction mainly through caspase-3-dependent epithelial apoptosis. Concomitant tight junction changes were caspase-independent. Anti-apoptotic and immune-modulatory substances appear to be promising agents for treatment of campylobacteriosis.
Collapse
Affiliation(s)
- Eduard Butkevych
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fábia Daniela Lobo de Sá
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Praveen Kumar Nattramilarasu
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
24
|
Schmidt AM, Escher U, Mousavi S, Tegtmeyer N, Boehm M, Backert S, Bereswill S, Heimesaat MM. Immunopathological properties of the Campylobacter jejuni flagellins and the adhesin CadF as assessed in a clinical murine infection model. Gut Pathog 2019; 11:24. [PMID: 31131028 PMCID: PMC6525468 DOI: 10.1186/s13099-019-0306-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/10/2019] [Indexed: 12/11/2022] Open
Abstract
Background Campylobacter jejuni infections constitute serious threats to human health with increasing prevalences worldwide. Our knowledge regarding the molecular mechanisms underlying host–pathogen interactions is still limited. Our group has established a clinical C. jejuni infection model based on abiotic IL-10−/− mice mimicking key features of human campylobacteriosis. In order to further validate this model for unraveling pathogen-host interactions mounting in acute disease, we here surveyed the immunopathological features of the important C. jejuni virulence factors FlaA and FlaB and the major adhesin CadF (Campylobacter adhesin to fibronectin), which play a role in bacterial motility, protein secretion and adhesion, respectively. Methods and results Therefore, abiotic IL-10−/− mice were perorally infected with C. jejuni strain 81-176 (WT) or with its isogenic flaA/B (ΔflaA/B) or cadF (ΔcadF) deletion mutants. Cultural analyses revealed that WT and ΔcadF but not ΔflaA/B bacteria stably colonized the stomach, duodenum and ileum, whereas all three strains were present in the colon at comparably high loads on day 6 post-infection. Remarkably, despite high colonic colonization densities, murine infection with the ΔflaA/B strain did not result in overt campylobacteriosis, whereas mice infected with ΔcadF or WT were suffering from acute enterocolitis at day 6 post-infection. These symptoms coincided with pronounced pro-inflammatory immune responses, not only in the intestinal tract, but also in other organs such as the liver and kidneys and were accompanied with systemic inflammatory responses as indicated by increased serum MCP-1 concentrations following C. jejuni ΔcadF or WT, but not ΔflaA/B strain infection. Conclusion For the first time, our observations revealed that the C. jejuni flagellins A/B, but not adhesion mediated by CadF, are essential for inducing murine campylobacteriosis. Furthermore, the secondary abiotic IL-10−/− infection model has been proven suitable not only for detailed investigations of immunological aspects of campylobacteriosis, but also for differential analyses of the roles of distinct C. jejuni virulence factors in induction and progression of disease. Electronic supplementary material The online version of this article (10.1186/s13099-019-0306-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna-Maria Schmidt
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Ulrike Escher
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Soraya Mousavi
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Nicole Tegtmeyer
- 2Institute for Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg, Erlangen, Germany
| | - Manja Boehm
- 2Institute for Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg, Erlangen, Germany
| | - Steffen Backert
- 2Institute for Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg, Erlangen, Germany
| | - Stefan Bereswill
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| | - Markus M Heimesaat
- Department of Microbiology, Institute of Microbiology, Infectious Diseases and Immunology, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, CC5, Campus Benjamin Franklin, FEM, Garystr. 5, 14195 Berlin, Germany
| |
Collapse
|
25
|
Harrer A, Bücker R, Boehm M, Zarzecka U, Tegtmeyer N, Sticht H, Schulzke JD, Backert S. Campylobacter jejuni enters gut epithelial cells and impairs intestinal barrier function through cleavage of occludin by serine protease HtrA. Gut Pathog 2019; 11:4. [PMID: 30805031 PMCID: PMC6373145 DOI: 10.1186/s13099-019-0283-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/01/2019] [Indexed: 01/17/2023] Open
Abstract
Campylobacter jejuni secretes HtrA (high temperature requirement protein A), a serine protease that is involved in virulence. Here, we investigated the interaction of HtrA with the host protein occludin, a tight junction strand component. Immunofluorescence studies demonstrated that infection of polarized intestinal Caco-2 cells with C. jejuni strain 81-176 resulted in a redistribution of occludin away from the tight junctions into the cytoplasm, an effect that was also observed in human biopsies during acute campylobacteriosis. Occludin knockout Caco-2 cells were generated by CRISPR/Cas9 technology. Inactivation of this gene affected the polarization of the cells in monolayers and transepithelial electrical resistance (TER) was reduced, compared to wild-type Caco-2 cells. Although tight junctions were still being formed, occludin deficiency resulted in a slight decrease of the tight junction plaque protein ZO-1, which was redistributed off the tight junction into the lateral plasma membrane. Adherence of C. jejuni to Caco-2 cell monolayers was similar between the occludin knockout compared to wild-type cells, but invasion was enhanced, indicating that deletion of occludin allowed larger numbers of bacteria to pass the tight junctions and to reach basal membranes to target the fibronectin receptor followed by cell entry. Finally, we discovered that purified C. jejuni HtrA cleaves recombinant occludin in vitro to release a 37 kDa carboxy-terminal fragment. The same cleavage fragment was observed in Western blots upon infection of polarized Caco-2 cells with wild-type C. jejuni, but not with isogenic ΔhtrA mutants. HtrA cleavage was mapped to the second extracellular loop of occludin, and a putative cleavage site was identified. In conclusion, HtrA functions as a secreted protease targeting the tight junctions, which enables the bacteria by cleaving occludin and subcellular redistribution of other tight junction proteins to transmigrate using a paracellular mechanism and subsequently invade epithelial cells.
Collapse
Affiliation(s)
- Aileen Harrer
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Roland Bücker
- 2Institut für Klinische Physiologie, Med. Klinik m.S. Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Manja Boehm
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Urszula Zarzecka
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany.,4Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, 80-308 Gdansk, Poland
| | - Nicole Tegtmeyer
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Heinrich Sticht
- 3Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jörg D Schulzke
- 2Institut für Klinische Physiologie, Med. Klinik m.S. Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Steffen Backert
- 1Division of Microbiology, Dept. of Biology, University of Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| |
Collapse
|
26
|
Host cellular unfolded protein response signaling regulates Campylobacter jejuni invasion. PLoS One 2018; 13:e0205865. [PMID: 30321237 PMCID: PMC6188877 DOI: 10.1371/journal.pone.0205865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/02/2018] [Indexed: 12/20/2022] Open
Abstract
Campylobacter jejuni is a major cause of bacterial foodborne illness in humans worldwide. Bacterial entry into a host eukaryotic cell involves the initial steps of adherence and invasion, which generally activate several cell-signaling pathways that induce the activation of innate defense systems, which leads to the release of proinflammatory cytokines and induction of apoptosis. Recent studies have reported that the unfolded protein response (UPR), a system to clear unfolded proteins from the endoplasmic reticulum (ER), also participates in the activation of cellular defense mechanisms in response to bacterial infection. However, no study has yet investigated the role of UPR in C. jejuni infection. Hence, the aim of this study was to deduce the role of UPR signaling via induction of ER stress in the process of C. jejuni infection. The results suggest that C. jejuni infection suppresses global protein translation. Also, 12 h of C. jejuni infection induced activation of the eIF2α pathway and expression of the transcription factor CHOP. Interestingly, bacterial invasion was facilitated by knockdown of UPR-associated signaling factors and treatment with the ER stress inducers, thapsigargin and tunicamycin, decreased the invasive ability of C. jejuni. An investigation into the mechanism of UPR-mediated inhibition of C. jejuni invasion showed that UPR signaling did not affect bacterial adhesion to or survival in the host cells. Further, Salmonella Enteritidis or FITC-dextran intake were not regulated by UPR signaling. These results indicated that the effect of UPR on intracellular intake was specifically found in C. jejuni infection. These findings are the first to describe the role of UPR in C. jejuni infection and revealed the participation of a new signaling pathway in C. jejuni invasion. UPR signaling is involved in defense against the early step of C. jejuni invasion and thus presents a potential therapeutic target for the treatment of C. jejuni infection.
Collapse
|
27
|
Boehm M, Simson D, Escher U, Schmidt AM, Bereswill S, Tegtmeyer N, Backert S, Heimesaat MM. Function of Serine Protease HtrA in the Lifecycle of the Foodborne Pathogen Campylobacter jejuni. Eur J Microbiol Immunol (Bp) 2018; 8:70-77. [PMID: 30345086 PMCID: PMC6186014 DOI: 10.1556/1886.2018.00011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022] Open
Abstract
Campylobacter jejuni is a major food-borne zoonotic pathogen, responsible for a large proportion of bacterial gastroenteritis cases, as well as Guillian-Barré and Miller-Fisher syndromes. During infection, tissue damage is mainly caused by bacteria invading epithelial cells and traversing the intestinal barrier. C. jejuni is able to enter the lamina propria and the bloodstream and may move into other organs, such as spleen, liver, or mesenteric lymph nodes. However, the involved molecular mechanisms are not fully understood. C. jejuni can transmigrate effectively across polarized intestinal epithelial cells mainly by the paracellular route using the serine protease high-temperature requirement A (HtrA). However, it appears that HtrA has a dual function, as it also acts as a chaperone, interacting with denatured or misfolded periplasmic proteins under stress conditions. Here, we review recent progress on the role of HtrA in C. jejuni pathogenesis. HtrA can be transported into the extracellular space and cleaves cell-to-cell junction factors, such as E-cadherin and probably others, disrupting the epithelial barrier and enabling paracellular transmigration of the bacteria. The secretion of HtrA is a newly discovered strategy also utilized by other pathogens. Thus, secreted HtrA proteases represent highly attractive targets for anti-bacterial treatment and may provide a suitable candidate for vaccine development.
Collapse
Affiliation(s)
- Manja Boehm
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen/Nuremberg, Staudtstr. 5, D-91058 Erlangen, Germany
| | - Daniel Simson
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen/Nuremberg, Staudtstr. 5, D-91058 Erlangen, Germany
| | - Ulrike Escher
- Department of Microbiology and Infection Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Anna-Maria Schmidt
- Department of Microbiology and Infection Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Department of Microbiology and Infection Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen/Nuremberg, Staudtstr. 5, D-91058 Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen/Nuremberg, Staudtstr. 5, D-91058 Erlangen, Germany
| | - Markus M Heimesaat
- Department of Microbiology and Infection Immunology, Charité - University Medicine Berlin, Berlin, Germany
| |
Collapse
|
28
|
Hatayama S, Shimohata T, Amano S, Kido J, Nguyen AQ, Sato Y, Kanda Y, Tentaku A, Fukushima S, Nakahashi M, Uebanso T, Mawatari K, Takahashi A. Cellular Tight Junctions Prevent Effective Campylobacter jejuni Invasion and Inflammatory Barrier Disruption Promoting Bacterial Invasion from Lateral Membrane in Polarized Intestinal Epithelial Cells. Front Cell Infect Microbiol 2018; 8:15. [PMID: 29441328 PMCID: PMC5797580 DOI: 10.3389/fcimb.2018.00015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/12/2018] [Indexed: 01/23/2023] Open
Abstract
Campylobacter jejuni invasion is closely related to C. jejuni pathogenicity. The intestinal epithelium contains polarized epithelial cells that form tight junctions (TJs) to provide a physical barrier against bacterial invasion. Previous studies indicated that C. jejuni invasion of non-polarized cells involves several cellular features, including lipid rafts. However, the dynamics of C. jejuni invasion of polarized epithelial cells are not fully understood. Here we investigated the interaction between C. jejuni invasion and TJ formation to characterize the mechanism of C. jejuni invasion in polarized epithelial cells. In contrast to non-polarized epithelial cells, C. jejuni invasion was not affected by depletion of lipid rafts in polarized epithelial cells. However, depletion of lipid rafts significantly decreased C. jejuni invasion in TJ disrupted cells or basolateral infection and repair of cellular TJs suppressed lipid raft-mediated C. jejuni invasion in polarized epithelial cells. In addition, pro-inflammatory cytokine, TNF-α treatment that induce TJ disruption promote C. jejuni invasion and lipid rafts depletion significantly reduced C. jejuni invasion in TNF-α treated cells. These data demonstrated that TJs prevent C. jejuni invasion from the lateral side of epithelial cells, where they play a main part in bacterial invasion and suggest that C. jejuni invasion could be increased in inflammatory condition. Therefore, maintenance of TJs integrity should be considered important in the development of novel therapies for C. jejuni infection.
Collapse
Affiliation(s)
- Sho Hatayama
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Takaaki Shimohata
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Sachie Amano
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Junko Kido
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Anh Q Nguyen
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Yuri Sato
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Yuna Kanda
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Aya Tentaku
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Shiho Fukushima
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Mutsumi Nakahashi
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Takashi Uebanso
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Kazuaki Mawatari
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| | - Akira Takahashi
- Department of Preventive Environment and Nutrition, Institute of Biomedical Science, Tokushima University, Tokushima, Japan
| |
Collapse
|
29
|
Identification and differentiation of Campylobacter species by high-resolution melting curve analysis. Microb Pathog 2017; 108:109-113. [DOI: 10.1016/j.micpath.2017.05.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 04/28/2017] [Accepted: 05/02/2017] [Indexed: 11/24/2022]
|
30
|
Ayllón N, Jiménez-Marín Á, Argüello H, Zaldívar-López S, Villar M, Aguilar C, Moreno A, De La Fuente J, Garrido JJ. Comparative Proteomics Reveals Differences in Host-Pathogen Interaction between Infectious and Commensal Relationship with Campylobacter jejuni. Front Cell Infect Microbiol 2017; 7:145. [PMID: 28491823 PMCID: PMC5405767 DOI: 10.3389/fcimb.2017.00145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/07/2017] [Indexed: 02/04/2023] Open
Abstract
Campylobacter jejuni is the leading food-borne poisoning in industrialized countries. While the bacteria causes disease in humans, it merely colonizes the gut in poultry or pigs, where seems to establish a commensal relationship. Until now, few studies have been conducted to elucidate the relationship between C. jejuni and its different hosts. In this work, a comparative proteomics approach was used to identify the underlying mechanisms involved in the divergent outcome following C. jejuni infection in human and porcine host. Human (INT-407) and porcine (IPEC-1) intestinal cell lines were infected by C. jejuni for 3 h (T3h) and 24 h (T24h). C. jejuni infection prompted an intense inflammatory response at T3h in human intestinal cells, mainly characterized by expression of proteins involved in cell spreading, cell migration and promotion of reactive oxygen species (ROS). Proteomic analysis evidenced significantly regulated biofunctions in human cells related with engulfment and endocytosis, and supported by canonical pathways associated to infection such as caveolar- and clathrin-mediated endocytosis signaling. In porcine IPEC-1 cells, inflammatory response as well as signaling pathways that control cellular functions such as cell migration, endocytosis and cell cycle progression resulted downregulated. These differences in the host response to infection were supported by the different pattern of adhesion and invasion proteins expressed by C. jejuni in human and porcine cells. No marked differences in expression of virulence factors involved in adaptive response and iron acquisition functions were observed. Therefore, the results of this study suggest that both host and pathogen factors are responsible for commensal or infectious character of C. jejuni in different hosts.
Collapse
Affiliation(s)
- Nieves Ayllón
- SaBio, Instituto de Investigación en Recursos Cinegéticos (CSIC-UCLM-JCCM)Ciudad Real, Spain
| | - Ángeles Jiménez-Marín
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de CórdobaCórdoba, Spain
| | - Héctor Argüello
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de CórdobaCórdoba, Spain
| | - Sara Zaldívar-López
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de CórdobaCórdoba, Spain
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos (CSIC-UCLM-JCCM)Ciudad Real, Spain
| | - Carmen Aguilar
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de CórdobaCórdoba, Spain
| | - Angela Moreno
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de CórdobaCórdoba, Spain
| | - José De La Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos (CSIC-UCLM-JCCM)Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State UniversityStillwater, OK, USA
| | - Juan J. Garrido
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de CórdobaCórdoba, Spain
| |
Collapse
|
31
|
Freitag CM, Strijbis K, van Putten JPM. Host cell binding of the flagellar tip protein of Campylobacter jejuni. Cell Microbiol 2017; 19. [PMID: 28008697 DOI: 10.1111/cmi.12714] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 12/16/2016] [Indexed: 12/29/2022]
Abstract
Flagella are nanofibers that drive bacterial movement. The filaments are generally composed of thousands of tightly packed flagellin subunits with a terminal cap protein, named FliD. Here, we report that the FliD protein of the bacterial pathogen Campylobacter jejuni binds to host cells. Live-cell imaging and confocal microscopy showed initial contact of the bacteria with epithelial cells via the flagella tip. Recombinant FliD protein bound to the surface of intestinal epithelial cells in a dose-dependent fashion. Search for the FliD binding site on the host cell using cells with defined glycosylation defects indicated glycosaminoglycans as a putative target. Heparinase treatment of wild type cells and an excess of soluble heparin abolished FliD binding. Binding assays showed direct and specific binding of FliD to heparin. Addition of an excess of purified FliD or heparin reduced the attachment of viable C. jejuni to the host cells. The host cell binding domain of FliD was mapped to the central region of the protein. Overall, our results indicate that the C. jejuni flagellar tip protein FliD acts as an attachment factor that interacts with cell surface heparan sulfate glycosaminoglycan receptors.
Collapse
Affiliation(s)
- Claudia M Freitag
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Karin Strijbis
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
32
|
Lee S, Lee J, Ha J, Choi Y, Kim S, Lee H, Yoon Y, Choi KH. Clinical relevance of infections with zoonotic and human oral species of Campylobacter. J Microbiol 2016; 54:459-67. [PMID: 27350611 DOI: 10.1007/s12275-016-6254-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/07/2016] [Accepted: 06/07/2016] [Indexed: 12/18/2022]
Abstract
Genus Campylobacter has been recognized as a causative bacterial agent of animal and human diseases. Human Campylobacter infections have caused more concern. Campylobacters can be classified into two groups in terms of their original host: zoonotic and human oral species. The major zoonotic species are Campylobacter jejuni and Campylobacter coli, which mostly reside in the intestines of avian species and are transmitted to humans via consumption of contaminated poultry products, thus causing human gastroenteritis and other diseases as sequelae. The other campylobacters, human oral species, include C. concisus, C. showae, C. gracilis, C. ureolyticus, C. curvus, and C. rectus. These species are isolated from the oral cavity, natural colonization site, but have potential clinical relevance in the periodontal region to varying extent. Two species, C. jejuni and C. coli, are believed to be mainly associated with intestinal diseases, but recent studies suggested that oral Campylobacter species also play a significant role in intestinal diseases. This review offers an outline of the two Campylobacter groups (zoonotic and human oral), their virulence traits, and the associated illnesses including gastroenteritis.
Collapse
Affiliation(s)
- Soomin Lee
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jeeyeon Lee
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jimyeong Ha
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Yukyung Choi
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Sejeong Kim
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Heeyoung Lee
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Yohan Yoon
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Kyoung-Hee Choi
- Department of Oral Microbiology, College of Dentistry, Wonkwang University, Iksan, Chonbuk, 54538, Republic of Korea.
| |
Collapse
|
33
|
Zhang Y, Feng H. Pathogenic effects of glucosyltransferase from Clostridium difficile toxins. Pathog Dis 2016; 74:ftw024. [PMID: 27044305 DOI: 10.1093/femspd/ftw024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2016] [Indexed: 01/13/2023] Open
Abstract
The glucosyltransferase domain ofClostridium difficiletoxins modifies guanine nucleotide-binding proteins of Rho family. It is the major virulent domain of the holotoxins. Various pathogenic effects ofC. difficiletoxins in response to Rho glucosylation have been investigated including cytoskeleton damage, cell death and inflammation. The most recent studies have revealed some significant characteristics of the holotoxins that are independent of glucosylating activity. These findings arouse discussion about the role of glucosyltransferase activity in toxin pathogenesis and open up new insights for toxin mechanism study. In this review, we summarize the pathogenic effects of glucosyltransferase domain of the toxins in the past years.
Collapse
Affiliation(s)
- Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland Baltimore, 650 W. Baltimore Street, Baltimore, MD 21201, USA
| |
Collapse
|
34
|
Bonfim-Melo A, Zanetti BF, Ferreira ÉR, Vandoninck S, Han SW, Van Lint J, Mortara RA, Bahia D. Trypanosoma cruziextracellular amastigotes trigger the protein kinase D1-cortactin-actin pathway during cell invasion. Cell Microbiol 2015; 17:1797-810. [DOI: 10.1111/cmi.12472] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 05/31/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Alexis Bonfim-Melo
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
| | - Bianca Ferrarini Zanetti
- Interdisciplinary Center for Gene Therapy (CINTERGEN); Universidade Federal de São Paulo (UNIFESP); São Paulo Brazil
| | - Éden Ramalho Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
| | - Sandy Vandoninck
- Department of Cellular and Molecular Medicine; University of Leuven; Leuven Belgium
| | - Sang Won Han
- Interdisciplinary Center for Gene Therapy (CINTERGEN); Universidade Federal de São Paulo (UNIFESP); São Paulo Brazil
| | - Johan Van Lint
- Department of Cellular and Molecular Medicine; University of Leuven; Leuven Belgium
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina; Universidade Federal de São Paulo (EPM-UNIFESP); São Paulo Brazil
- Departamento de Biologia Geral, Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais (ICB-UFMG); Belo Horizonte Brazil
| |
Collapse
|
35
|
Boehm M, Lind J, Backert S, Tegtmeyer N. Campylobacter jejuni serine protease HtrA plays an important role in heat tolerance, oxygen resistance, host cell adhesion, invasion, and transmigration. Eur J Microbiol Immunol (Bp) 2015; 5:68-80. [PMID: 25883795 DOI: 10.1556/eujmi-d-15-00003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/13/2015] [Indexed: 12/29/2022] Open
Abstract
Campylobacter jejuni is an important pathogen of foodborne illness. Transmigration across the intestinal epithelial barrier and invasion are considered as primary reasons for tissue damage triggered by C. jejuni. Using knockout mutants, it was shown that the serine protease HtrA may be important for stress tolerance and physiology of C. jejuni. HtrA is also secreted in the extra-cellular environment, where it can cleave junctional host cell proteins such as E-cadherin. Aim of the present study was to establish a genetic complementation system in two C. jejuni strains in order to introduce the wild-type htrA gene in trans, test known htrA phenotypes, and provide the basis to perform further mutagenesis. We confirm that reexpression of the htrA wild-type gene in ΔhtrA mutants restored the following phenotypes: 1) C. jejuni growth at high temperature (44 °C), 2) growth under high oxygen stress conditions, 3) expression of proteolytically active HtrA oligomers, 4) secretion of HtrA into the supernatant, 5) cell attachment and invasion, and 6) transmigration across polarized epithelial cells. These results establish a genetic complementation system for htrA in C. jejuni, exclude polar effects in the ΔhtrA mutants, confirm important HtrA properties, and permit the discovery and dissection of new functions.
Collapse
Affiliation(s)
- Manja Boehm
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen/Nuremberg Staudtstr. 5, D-91058 Erlangen Germany
| | - Judith Lind
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen/Nuremberg Staudtstr. 5, D-91058 Erlangen Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen/Nuremberg Staudtstr. 5, D-91058 Erlangen Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen/Nuremberg Staudtstr. 5, D-91058 Erlangen Germany
| |
Collapse
|
36
|
Alam J, Baek KJ, Choi YS, Kim YC, Choi Y. N-acetylcysteine and the human serum components that inhibit bacterial invasion of gingival epithelial cells prevent experimental periodontitis in mice. J Periodontal Implant Sci 2014; 44:266-73. [PMID: 25568806 PMCID: PMC4284374 DOI: 10.5051/jpis.2014.44.6.266] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/24/2014] [Indexed: 12/24/2022] Open
Abstract
PURPOSE We previously reported that human serum significantly reduces the invasion of various oral bacterial species into gingival epithelial cells in vitro. The aims of the present study were to characterize the serum component(s) responsible for the inhibition of bacterial invasion of epithelial cells and to examine their effect on periodontitis induced in mice. METHODS Immortalized human gingival epithelial (HOK-16B) cells were infected with various 5- (and 6-) carboxy-fluorescein diacetate succinimidyl ester-labeled oral bacteria, including Fusobacterium nucleatum, Provetella intermedia, Porphyromonas gingivalis, and Treponiema denticola, in the absence or presence of three major serum components (human serum albumin [HSA], pooled human IgG [phIgG] and α1-antitrypsin). Bacterial adhesion and invasion were determined by flow cytometry. The levels of intracellular reactive oxygen species (ROS) and activation of small GTPases were examined. Experimental periodontitis was induced by oral inoculation of P. gingivalis and T. denticola in Balb/c mice. RESULTS HSA and phIgG, but not α1-antitrypsin, efficiently inhibited the invasion of various oral bacterial species into HOK-16B cells. HSA but not phIgG decreased the adhesion of F. nucleatum onto host cells and the levels of intracellular ROS in HOK-16B cells. N-acetylcysteine (NAC), a ROS scavenger, decreased both the levels of intracellular ROS and invasion of F. nucleatum into HOK-16B cells, confirming the role of ROS in bacterial invasion. Infection with F. nucleatum activated Rac1, a regulator of actin cytoskeleton dynamics. Not only HSA and NAC but also phIgG decreased the F. nucleatum-induced activation of Rac1. Furthermore, both HSA plus phIgG and NAC significantly reduced the alveolar bone loss in the experimental periodontitis induced by P. gingivalis and T. denticola in mice. CONCLUSIONS NAC and the serum components HSA and phIgG, which inhibit bacterial invasion of oral epithelial cells in vitro, can successfully prevent experimental periodontitis.
Collapse
Affiliation(s)
- Jehan Alam
- Department of Oral Microbiology and Immunology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Keum Jin Baek
- Department of Oral Microbiology and Immunology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Yun Sik Choi
- Department of Oral Microbiology and Immunology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Yong Cheol Kim
- Department of Oral Microbiology and Immunology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| | - Youngnim Choi
- Department of Oral Microbiology and Immunology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Korea
| |
Collapse
|
37
|
Aguilar C, Jiménez-Marín Á, Martins RP, Garrido JJ. Interaction between Campylobacter and intestinal epithelial cells leads to a different proinflammatory response in human and porcine host. Vet Immunol Immunopathol 2014; 162:14-23. [DOI: 10.1016/j.vetimm.2014.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 09/02/2014] [Accepted: 09/16/2014] [Indexed: 10/24/2022]
|
38
|
Heimesaat MM, Alutis M, Grundmann U, Fischer A, Tegtmeyer N, Böhm M, Kühl AA, Göbel UB, Backert S, Bereswill S. The role of serine protease HtrA in acute ulcerative enterocolitis and extra-intestinal immune responses during Campylobacter jejuni infection of gnotobiotic IL-10 deficient mice. Front Cell Infect Microbiol 2014; 4:77. [PMID: 24959425 PMCID: PMC4050650 DOI: 10.3389/fcimb.2014.00077] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/22/2014] [Indexed: 12/24/2022] Open
Abstract
Campylobacter jejuni infections have a high prevalence worldwide and represent a significant socioeconomic burden. C. jejuni can cross the intestinal epithelial barrier as visualized in biopsies derived from human patients and animal models, however, the underlying molecular mechanisms and associated immunopathology are still not well understood. We have recently shown that the secreted serine protease HtrA (high temperature requirement A) plays a key role in C. jejuni cellular invasion and transmigration across polarized epithelial cells in vitro. In the present in vivo study we investigated the role of HtrA during C. jejuni infection of mice. We used the gnotobiotic IL-10−/− mouse model to study campylobacteriosis following peroral infection with the C. jejuni wild-type (WT) strain NCTC11168 and the isogenic, non-polar NCTC11168ΔhtrA deletion mutant. Six days post infection (p.i.) with either strain mice harbored comparable intestinal C. jejuni loads, whereas ulcerative enterocolitis was less pronounced in mice infected with the ΔhtrA mutant strain. Moreover, ΔhtrA mutant infected mice displayed lower apoptotic cell numbers in the large intestinal mucosa, less colonic accumulation of neutrophils, macrophages and monocytes, lower large intestinal nitric oxide, IFN-γ, and IL-6 as well as lower TNF-α and IL-6 serum concentrations as compared to WT strain infected mice at day 6 p.i. Notably, immunopathological responses were not restricted to the intestinal tract given that liver and kidneys exhibited mild histopathological changes 6 days p.i. with either C. jejuni strain. We also found that hepatic and renal nitric oxide levels or renal TNF-α concentrations were lower in the ΔhtrA mutant as compared to WT strain infected mice. In conclusion, we show here that the C. jejuni HtrA protein plays a pivotal role in inducing host cell apoptosis and immunopathology during murine campylobacteriosis in the gut in vivo.
Collapse
Affiliation(s)
- Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin Berlin, Germany
| | - Marie Alutis
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin Berlin, Germany
| | - Ursula Grundmann
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin Berlin, Germany
| | - André Fischer
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin Berlin, Germany
| | - Nicole Tegtmeyer
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg Erlangen, Germany
| | - Manja Böhm
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg Erlangen, Germany
| | - Anja A Kühl
- Department of Medicine I for Gastroenterology, Infectious Disease and Rheumatology/Research Center ImmunoSciences, Charité - University Medicine Berlin Berlin, Germany
| | - Ulf B Göbel
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin Berlin, Germany
| | - Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen/Nuremberg Erlangen, Germany
| | - Stefan Bereswill
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin Berlin, Germany
| |
Collapse
|
39
|
Heimesaat MM, Fischer A, Alutis M, Grundmann U, Boehm M, Tegtmeyer N, Göbel UB, Kühl AA, Bereswill S, Backert S. The impact of serine protease HtrA in apoptosis, intestinal immune responses and extra-intestinal histopathology during Campylobacter jejuni infection of infant mice. Gut Pathog 2014; 6:16. [PMID: 24883112 PMCID: PMC4040118 DOI: 10.1186/1757-4749-6-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 05/19/2014] [Indexed: 12/19/2022] Open
Abstract
Background Campylobacter jejuni has emerged as a leading cause of bacterial enterocolitis. The serine protease HtrA has been shown to be a pivotal, novel C. jejuni virulence factor involved in cell invasion and transmigration across polarised epithelial cells in vitro. However, the functional relevance of the htrA gene for the interaction of C. jejuni with the host immune system in the infant mouse infection model has not been investigated so far. Results Here we studied the role of C. jejuni htrA during infection of 3-weeks-old infant mice. Immediately after weaning, conventional wild-type mice were perorally infected with the NCTC11168∆htrA mutant (∆htrA) or the parental wild-type strain. Approximately one third of infected infant mice suffered from bloody diarrhea until day 7 post infection (p.i.), whereas colonic histopathological changes were rather moderate but comparable between the two strains. Interestingly, parental, but not ∆htrA mutant infected mice, displayed a multifold increase of apoptotic cells in the colonic mucosa at day 7 p.i., which was paralleled by higher colonic levels of pro-inflammatory cytokines such as TNF-α and IFN-γ and the matrix-degrading enzyme matrixmetalloproteinase-2 (MMP-2). Furthermore, higher numbers of proliferating cells could be observed in the colon of ∆htrA infected mice as compared to the parental wild-type strain. Remarkably, as early as 7 days p.i. infant mice also exhibited inflammatory changes in extra-intestinal compartments such as liver, kidneys and lungs, which were less distinct in kidneys and lungs following ∆htrA versus parental strain infection. However, live C. jejuni bacteria could not be found in these organs, suggesting the induction of systemic effects during intestinal infection. Conclusion Upon C. jejuni ∆htrA strain infection of infant mice, intestinal and extra-intestinal pro-inflammatory immune responses were ameliorated in the infant mouse model system. Future studies will shed further light onto the molecular mechanisms of host-pathogen interactions.
Collapse
Affiliation(s)
- Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 27, D-12203 Berlin, Germany
| | - André Fischer
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 27, D-12203 Berlin, Germany
| | - Marie Alutis
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 27, D-12203 Berlin, Germany
| | - Ursula Grundmann
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 27, D-12203 Berlin, Germany
| | - Manja Boehm
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen, Nuremberg, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen, Nuremberg, Germany
| | - Ulf B Göbel
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 27, D-12203 Berlin, Germany
| | - Anja A Kühl
- Department of Medicine I for Gastroenterology, Infectious Disease and Rheumatology / Research Center ImmunoSciences (RCIS), Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 27, D-12203 Berlin, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen, Nuremberg, Germany
| |
Collapse
|
40
|
Popoff MR. Bacterial factors exploit eukaryotic Rho GTPase signaling cascades to promote invasion and proliferation within their host. Small GTPases 2014; 5:28209. [PMID: 25203748 DOI: 10.4161/sgtp.28209] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Actin cytoskeleton is a main target of many bacterial pathogens. Among the multiple regulation steps of the actin cytoskeleton, bacterial factors interact preferentially with RhoGTPases. Pathogens secrete either toxins which diffuse in the surrounding environment, or directly inject virulence factors into target cells. Bacterial toxins, which interfere with RhoGTPases, and to some extent with RasGTPases, catalyze a covalent modification (ADPribosylation, glucosylation, deamidation, adenylation, proteolysis) blocking these molecules in their active or inactive state, resulting in alteration of epithelial and/or endothelial barriers, which contributes to dissemination of bacteria in the host. Injected bacterial virulence factors preferentially manipulate the RhoGTPase signaling cascade by mimicry of eukaryotic regulatory proteins leading to local actin cytoskeleton rearrangement, which mediates bacterial entry into host cells or in contrast escape to phagocytosis and immune defense. Invasive bacteria can also manipulate RhoGTPase signaling through recognition and stimulation of cell surface receptor(s). Changes in RhoGTPase activation state is sensed by the innate immunity pathways and allows the host cell to adapt an appropriate defense response.
Collapse
Affiliation(s)
- Michel R Popoff
- Unité des Bactéries anaérobies et Toxines; Institut Pasteur; Paris, France
| |
Collapse
|
41
|
Di Giannatale E, Di Serafino G, Zilli K, Alessiani A, Sacchini L, Garofolo G, Aprea G, Marotta F. Characterization of antimicrobial resistance patterns and detection of virulence genes in Campylobacter isolates in Italy. SENSORS 2014; 14:3308-22. [PMID: 24556669 PMCID: PMC3958300 DOI: 10.3390/s140203308] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 02/12/2014] [Accepted: 02/14/2014] [Indexed: 01/20/2023]
Abstract
Campylobacter has developed resistance to several antimicrobial agents over the years, including macrolides, quinolones and fluoroquinolones, becoming a significant public health hazard. A total of 145 strains derived from raw milk, chicken faeces, chicken carcasses, cattle faeces and human faeces collected from various Italian regions, were screened for antimicrobial susceptibility, molecular characterization (SmaI pulsed-field gel electrophoresis) and detection of virulence genes (sequencing and DNA microarray analysis). The prevalence of C. jejuni and C. coli was 62.75% and 37.24% respectively. Antimicrobial susceptibility revealed a high level of resistance for ciprofloxacin (62.76%), tetracycline (55.86%) and nalidixic acid (55.17%). Genotyping of Campylobacter isolates using PFGE revealed a total of 86 unique SmaI patterns. Virulence gene profiles were determined using a new microbial diagnostic microarray composed of 70-mer oligonucleotide probes targeting genes implicated in Campylobacter pathogenicity. Correspondence between PFGE and microarray clusters was observed. Comparisons of PFGE and virulence profiles reflected the high genetic diversity of the strains examined, leading us to speculate different degrees of pathogenicity inside Campylobacter populations.
Collapse
Affiliation(s)
| | | | - Katiuscia Zilli
- LNR Campylobacter, IZSAM G.Caporale, via Campo Boario 64100 Teramo, Italy.
| | | | - Lorena Sacchini
- LNR Campylobacter, IZSAM G.Caporale, via Campo Boario 64100 Teramo, Italy.
| | - Giuliano Garofolo
- LNR Campylobacter, IZSAM G.Caporale, via Campo Boario 64100 Teramo, Italy.
| | - Giuseppe Aprea
- LNR Campylobacter, IZSAM G.Caporale, via Campo Boario 64100 Teramo, Italy.
| | - Francesca Marotta
- LNR Campylobacter, IZSAM G.Caporale, via Campo Boario 64100 Teramo, Italy.
| |
Collapse
|
42
|
Konkel ME, Samuelson DR, Eucker TP, Shelden EA, O'Loughlin JL. Invasion of epithelial cells by Campylobacter jejuni is independent of caveolae. Cell Commun Signal 2013; 11:100. [PMID: 24364863 PMCID: PMC3880046 DOI: 10.1186/1478-811x-11-100] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 12/17/2013] [Indexed: 11/20/2022] Open
Abstract
Caveolae are 25–100 nm flask-like membrane structures enriched in cholesterol and glycosphingolipids. Researchers have proposed that Campylobacter jejuni require caveolae for cell invasion based on the finding that treatment of cells with the cholesterol-depleting compounds filipin III or methyl-β-cyclodextrin (MβCD) block bacterial internalization in a dose-dependent manner. The purpose of this study was to determine the role of caveolae and caveolin-1, a principal component of caveolae, in C. jejuni internalization. Consistent with previous work, we found that the treatment of HeLa cells with MβCD inhibited C. jejuni internalization. However, we also found that the treatment of HeLa cells with caveolin-1 siRNA, which resulted in greater than a 90% knockdown in caveolin-1 protein levels, had no effect on C. jejuni internalization. Based on this observation we performed a series of experiments that demonstrate that MβCD acts broadly, disrupting host cell lipid rafts and C. jejuni-induced cell signaling. More specifically, we found that MβCD inhibits the cellular events necessary for C. jejuni internalization, including membrane ruffling and Rac1 GTPase activation. We also demonstrate that MβCD disrupted the association of the β1 integrin and EGF receptor, which are required for the maximal invasion of epithelial cells. In agreement with these findings, C. jejuni were able to invade human Caco-2 cells, which are devoid of caveolae, at a level equal to that of HeLa cells. Taken together, the results of our study demonstrate that C. jejuni internalization occurs in a caveolae-independent manner.
Collapse
Affiliation(s)
- Michael E Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Life Sciences Bldg, Room 302c, Pullman, WA, USA.
| | | | | | | | | |
Collapse
|
43
|
Tareen AM, Lüder CGK, Zautner AE, Groß U, Heimesaat MM, Bereswill S, Lugert R. The Campylobacter jejuni Cj0268c protein is required for adhesion and invasion in vitro. PLoS One 2013; 8:e81069. [PMID: 24303031 PMCID: PMC3841222 DOI: 10.1371/journal.pone.0081069] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/09/2013] [Indexed: 12/24/2022] Open
Abstract
Adherence of Campylobacter jejuni to its particular host cells is mediated by several pathogen proteins. We screened a transposon-based mutant library of C. jejuni in order to identify clones with an invasion deficient phenotype towards Caco2 cells and detected a mutant with the transposon insertion in gene cj0268c. In vitro characterization of a generated non-random mutant, the mutant complemented with an intact copy of cj0268c and parental strain NCTC 11168 confirmed the relevance of Cj0268c in the invasion process, in particular regarding adherence to host cells. Whereas Cj0268c does not impact autoagglutination or motility of C. jejuni, heterologous expression in E. coli strain DH5α enhanced the potential of the complemented E. coli strain to adhere to Caco2 cells significantly and, thus, indicates that Cj0268c does not need to interact with other C. jejuni proteins to develop its adherence-mediating phenotype. Flow cytometric measurements of E. coli expressing Cj0268c indicate a localization of the protein in the periplasmic space with no access of its C-terminus to the bacterial surface. Since a respective knockout mutant possesses clearly reduced resistance to Triton X-100 treatment, Cj0268c contributes to the stability of the bacterial cell wall. Finally, we could show that the presence of cj0268c seems to be ubiquitous in isolates of C. jejuni and does not correlate with specific clonal groups regarding pathogenicity or pathogen metabolism.
Collapse
Affiliation(s)
- A. Malik Tareen
- University Medical Center Göttingen, Institute for Medical Microbiology, Göttingen, Germany
| | - Carsten G. K. Lüder
- University Medical Center Göttingen, Institute for Medical Microbiology, Göttingen, Germany
| | - Andreas E. Zautner
- University Medical Center Göttingen, Institute for Medical Microbiology, Göttingen, Germany
- University Medical Center Göttingen, Department of Clinical Chemistry/Central Laboratory, Göttingen, Germany
| | - Uwe Groß
- University Medical Center Göttingen, Institute for Medical Microbiology, Göttingen, Germany
| | - Markus M. Heimesaat
- Charité – University Medicine Berlin, Department of Microbiology and Hygiene, Berlin, Germany
| | - Stefan Bereswill
- Charité – University Medicine Berlin, Department of Microbiology and Hygiene, Berlin, Germany
| | - Raimond Lugert
- University Medical Center Göttingen, Institute for Medical Microbiology, Göttingen, Germany
- * E-mail:
| |
Collapse
|
44
|
Samuelson DR, Konkel ME. Serine phosphorylation of cortactin is required for maximal host cell invasion by Campylobacter jejuni. Cell Commun Signal 2013; 11:82. [PMID: 24188565 PMCID: PMC3832248 DOI: 10.1186/1478-811x-11-82] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 10/23/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Campylobacter jejuni causes acute disease characterized by severe diarrhea containing blood and leukocytes, fever, and abdominal cramping. Disease caused by C. jejuni is dependent on numerous bacterial and host factors. C. jejuni invasion of the intestinal epithelial cells is seen in both clinical samples and animal models indicating that host cell invasion is, in part, necessary for disease. C. jejuni utilizes a flagellar Type III Secretion System (T3SS) to deliver the Campylobacter invasion antigens (Cia) to host cells. The Cia proteins modulate host cell signaling leading to actin cytoskeleton rearrangement necessary for C. jejuni host cell invasion, and are required for the development of disease. RESULTS This study was based on the hypothesis that the C. jejuni CiaD effector protein mediates Erk 1/2 dependent cytoskeleton rearrangement. We showed that CiaD was required for the maximal phosphorylation of Erk 1/2 by performing an immunoblot with a p-Erk 1/2 specific antibody and that Erk 1/2 participates in C. jejuni invasion of host cells by performing the gentamicin protection assay in the presence and absence of the PD98059 (a potent inhibitor of Erk 1/2 activation). CiaD was also found to be required for the maximal phosphorylation of cortactin S405 and S418, as judged by immunoblot analysis. The response of human INT 407 epithelial cells to infection with C. jejuni was evaluated by confocal microscopy and scanning electron microscopy to determine the extent of membrane ruffling. This analysis revealed that CiaD, Erk 1/2, and cortactin participate in C. jejuni-induced membrane ruffling. Finally, cortactin and N-WASP were found to be involved in C. jejuni invasion of host cells using siRNA to N-WASP, and siRNA to cortactin, coupled with the gentamicin protection assay. CONCLUSION We conclude that CiaD is involved in the activation of Erk 1/2 and that activated Erk 1/2 facilitates C. jejuni invasion by phosphorylation of cortactin on serine 405 and 418. This is the first time that cortactin and N-WASP have been shown to be involved in C. jejuni invasion of host cells. These data also provide a mechanistic basis for the requirement of Erk 1/2 in C. jejuni-mediated cytoskeletal rearrangement.
Collapse
Affiliation(s)
| | - Michael E Konkel
- School of Molecular Biosciences, Washington State University, College of Veterinary Medicine, Life Sciences Bldg, Room 302c, Pullman, Washington 99164-7520, USA.
| |
Collapse
|
45
|
Citalán-Madrid AF, García-Ponce A, Vargas-Robles H, Betanzos A, Schnoor M. Small GTPases of the Ras superfamily regulate intestinal epithelial homeostasis and barrier function via common and unique mechanisms. Tissue Barriers 2013; 1:e26938. [PMID: 24868497 PMCID: PMC3942330 DOI: 10.4161/tisb.26938] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/21/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022] Open
Abstract
The intestinal epithelium forms a stable barrier protecting underlying tissues from pathogens in the gut lumen. This is achieved by specialized integral membrane structures such as tight and adherens junctions that connect neighboring cells and provide stabilizing links to the cytoskeleton. Junctions are constantly remodeled to respond to extracellular stimuli. Assembly and disassembly of junctions is regulated by interplay of actin remodeling, endocytotic recycling of junctional proteins, and various signaling pathways. Accumulating evidence implicate small G proteins of the Ras superfamily as important signaling molecules for the regulation of epithelial junctions. They function as molecular switches circling between an inactive GDP-bound and an active GTP-bound state. Once activated, they bind different effector molecules to control cellular processes required for correct junction assembly, maintenance and remodelling. Here, we review recent advances in understanding how GTPases of the Rho, Ras, Rab and Arf families contribute to intestinal epithelial homeostasis.
Collapse
Affiliation(s)
- Alí Francisco Citalán-Madrid
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Alexander García-Ponce
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Hilda Vargas-Robles
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Abigail Betanzos
- Department of Infectomics and Molecular Pathogenesis; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| |
Collapse
|
46
|
The fibronectin-binding motif within FlpA facilitates Campylobacter jejuni adherence to host cell and activation of host cell signaling. Emerg Microbes Infect 2013; 2:e65. [PMID: 26038437 PMCID: PMC3826066 DOI: 10.1038/emi.2013.65] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 11/12/2022]
Abstract
Campylobacter jejuni is a gram-negative, curved and rod-shaped bacterium that causes human gastroenteritis. Acute disease is associated with C. jejuni invasion of the intestinal epithelium. Epithelial cells infected with C. jejuni strains containing mutations in the FlpA and CadF fibronectin (Fn)-binding proteins exhibit reduced invasion of host cells and a C. jejuni CadF FlpA double mutant is impaired in the activation of epidermal growth factor receptor (EGFR) and Rho GTPase Rac1. Although these observations establish a role for Fn-binding proteins during C. jejuni invasion, their mechanistic contributions to invasion-associated signaling are unclear. We examined FlpA, a C. jejuni Fn-binding protein composed of three FNIII-like repeats D1, D2 and D3, to identify the interactions required for cellular adherence on pathogen-induced host cell signaling. We report that FlpA binds the Fn gelatin-binding domain via a motif within the D2 repeat. Epithelial cells infected with a flpA mutant exhibited decreased Rac1 activation and reduced membrane ruffling that coincided with impaired delivery of the secreted Cia proteins and reduced cell association. Phosphorylation of the Erk1/2 kinase, a downstream effector of EGFR signaling, was specifically associated with FlpA-mediated activation of β1-integrin and EGFR signaling. In vivo experiments revealed that FlpA is necessary for C. jejuni disease based on bacterial dissemination to the spleen of IL-10−/− germ-free mice. Thus, a novel Fn-binding motif within FlpA potentiates activation of Erk1/2 signaling via β1-integrin during C. jejuni infection.
Collapse
|
47
|
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]
|
48
|
Backert S, Boehm M, Wessler S, Tegtmeyer N. Transmigration route of Campylobacter jejuni across polarized intestinal epithelial cells: paracellular, transcellular or both? Cell Commun Signal 2013; 11:72. [PMID: 24079544 PMCID: PMC3850506 DOI: 10.1186/1478-811x-11-72] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 09/18/2013] [Indexed: 02/08/2023] Open
Abstract
Intact intercellular junctions and cellular matrix contacts are crucial structural components for the formation and maintenance of epithelial barrier functions in humans to control the commensal flora and protect against intruding microbes. Campylobacter jejuni is one of the most important zoonotic pathogens causing food-borne gastroenteritis and potentially more severe diseases such as reactive arthritis or Guillain–Barré syndrome. Crossing the intestinal epithelial barrier and host cell invasion by C. jejuni are considered to represent the primary reasons of gut tissue damage in humans and various animal model systems including monkeys, piglets, rabbits, hamsters and ferrets. C. jejuni is also able to invade underlying tissues such as the lamina propria, can enter the bloodstream, and possibly reach distinct organs such as spleen, liver or mesenteric lymph nodes. However, the molecular mechanisms as well as major bacterial and host cell factors involved in these activities are poorly understood. Various models exist by which the pathogen can trigger its own transmigration across polarized intestinal epithelial cells in vitro, the paracellular and/or transcellular mechanism. Recent studies suggest that bacterial factors such as flagellum, serine protease HtrA and lipooligosaccharide LOS may play an active role in bacterial transmigration. Here we review our knowledge on transmigration of C. jejuni as well as some other Campylobacter species, and discuss the pros and cons for the route(s) taken to travel across polarized epithelial cell monolayers. These studies provide fresh insights into the infection strategies employed by this important pathogen.
Collapse
Affiliation(s)
- Steffen Backert
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen/Nuremberg, Staudtstr, 5, D-91058, Erlangen, Germany.
| | | | | | | |
Collapse
|
49
|
Boehm M, Haenel I, Hoy B, Brøndsted L, Smith TG, Hoover T, Wessler S, Tegtmeyer N. Extracellular secretion of protease HtrA from Campylobacter jejuni is highly efficient and independent of its protease activity and flagellum. Eur J Microbiol Immunol (Bp) 2013; 3:163-73. [PMID: 24265934 DOI: 10.1556/eujmi.3.2013.3.3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 05/13/2013] [Indexed: 01/01/2023] Open
Abstract
The serine protease HtrA of C. jejuni has been identified as a novel secreted virulence factor which opens cell-to-cell junctions by cleaving E-cadherin. Efficient C. jejuni transmigration across polarized human epithelial cells requires the intact flagellum and HtrA; however, the mechanism of HtrA secretion into the supernatant is unknown. Here we show that HtrA secretion is highly efficient and does not require its proteolytic activity because the protease-inactive S197A mutant is secreted like wild-type HtrA. In addition, the flagellar mutants ΔflaA/B, ΔfliI, ΔflgH, ΔflhA, ΔflhB, and ΔflgS were also able to secrete HtrA in high amounts, while they were strongly attenuated in secreting the well-known invasion antigen CiaB. We also tested several culture media and cell lines of different origin such as human, mouse, hamster, dog, and chicken for their ability to influence HtrA secretion. Interestingly, HtrA was effectively secreted in the presence of most but not all cell lines and media, albeit at different levels, but secretion was significantly higher when fetal calf serum (FCS) was added. These results demonstrate that HtrA secretion by Campylobacter proceeds independent of HtrA's protease activity, the flagellum and origin of cell lines, but can be strongly enhanced by molecular compound(s) present in FCS.
Collapse
|
50
|
Bullman S, Lucid A, Corcoran D, Sleator RD, Lucey B. Genomic investigation into strain heterogeneity and pathogenic potential of the emerging gastrointestinal pathogen Campylobacter ureolyticus. PLoS One 2013; 8:e71515. [PMID: 24023611 PMCID: PMC3758288 DOI: 10.1371/journal.pone.0071515] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/01/2013] [Indexed: 12/29/2022] Open
Abstract
The recent detection and isolation of C. ureolyticus from patients with diarrhoeal illness and inflammatory bowel diseases warrants further investigation into its role as an emerging pathogen of the human gastrointestinal tract. Regarding the pathogenic mechanisms employed by this species we provide the first whole genome analysis of two C. ureolyticus isolates including the type strain. Comparative analysis, subtractive hybridisation and gene ontology searches against other Campylobacter species identifies the high degree of heterogenicity between C. ureolyticus isolates, in addition to the identification of 106 putative virulence associated factors, 52 of which are predicted to be secreted. Such factors encompass each of the known virulence tactics of pathogenic Campylobacter spp. including adhesion and colonisation (CadF, PEB1, IcmF and FlpA), invasion (ciaB and 16 virB-virD4 genes) and toxin production (S-layer RTX and ZOT). Herein, we provide the first virulence catalogue for C. ureolyticus, the components of which theoretically provide this emerging species with sufficient arsenal to establish pathology.
Collapse
Affiliation(s)
- Susan Bullman
- Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
| | - Alan Lucid
- Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
| | - Daniel Corcoran
- Department of Medical Microbiology, Cork University Hospital, Cork, Ireland
| | - Roy D. Sleator
- Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
- * E-mail:
| | - Brigid Lucey
- Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
- Department of Medical Microbiology, Cork University Hospital, Cork, Ireland
| |
Collapse
|