1
|
Zhang Y, Yang Z, Dai K, Hu B, Xu S, Wang Y, Lei L, Du S, Zhao Q, Huang X, Wu R, Yan Q, Wang Y, Cao S, Wen Y. Rab4b Promotes Cytolethal Distending Toxin from Glaesserella parasuis-Induced Cytotoxicity in PK-15 Cells. Toxins (Basel) 2024; 16:407. [PMID: 39330865 PMCID: PMC11435814 DOI: 10.3390/toxins16090407] [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: 08/26/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Glaesserella parasuis cytolethal distending toxin (GpCDT) can induce cell cycle arrest and apoptosis. Our laboratory's previous work demonstrated that GTPase 4b (Rab4b) is a key host protein implicated in GpCDT-induced cytotoxicity. This study investigated the probable involvement of Rab4b in the process. Our study used CRISPR/Cas9 technology to create a Rab4b-knockout cell line. The results showed greater resistance to GpCDT-induced cell cytotoxicity. In contrast, forced Rab4b overexpression increased GpCDT-induced cytotoxicity. Further immunoprecipitation study reveals that GpCDT may bind with Rab4b. In PK-15 cells, GpCDT is transported to the early endosomes and late endosomes, while after knocking out Rab4b, GpCDT cannot be transported to the early endosome via vesicles. Rab4b appears essential for GpCDT-induced cytotoxicity in PK-15 cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.)
| |
Collapse
|
2
|
Wong Z, Ong EBB. Unravelling bacterial virulence factors in yeast: From identification to the elucidation of their mechanisms of action. Arch Microbiol 2024; 206:303. [PMID: 38878203 DOI: 10.1007/s00203-024-04023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Pathogenic bacteria employ virulence factors (VF) to establish infection and cause disease in their host. Yeasts, Saccharomyces cerevisiae and Saccharomyces pombe, are useful model organisms to study the functions of bacterial VFs and their interaction with targeted cellular processes because yeast processes and organelle structures are highly conserved and similar to higher eukaryotes. In this review, we describe the principles and applications of the yeast model for the identification and functional characterisation of bacterial VFs to investigate bacterial pathogenesis. The growth inhibition phenotype caused by the heterologous expression of bacterial VFs in yeast is commonly used to identify candidate VFs. Then, subcellular localisation patterns of bacterial VFs can provide further clues about their target molecules and functions during infection. Yeast knockout and overexpression libraries are also used to investigate VF interactions with conserved eukaryotic cell structures (e.g., cytoskeleton and plasma membrane), and cellular processes (e.g., vesicle trafficking, signalling pathways, and programmed cell death). In addition, the yeast growth inhibition phenotype is also useful for screening new drug leads that target and inhibit bacterial VFs. This review provides an updated overview of new tools, principles and applications to study bacterial VFs in yeast.
Collapse
Affiliation(s)
- ZhenPei Wong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800 USM, Malaysia
| | - Eugene Boon Beng Ong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800 USM, Malaysia.
| |
Collapse
|
3
|
Shenker BJ, Korostoff J, Walker LP, Zekavat A, Dhingra A, Kim TJ, Boesze-Battaglia K. Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin Induces Cellugyrin-(Synaptogyrin 2) Dependent Cellular Senescence in Oral Keratinocytes. Pathogens 2024; 13:155. [PMID: 38392893 PMCID: PMC10892517 DOI: 10.3390/pathogens13020155] [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: 12/20/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Recently, we reported that oral-epithelial cells (OE) are unique in their response to Aggregatibacter actinomycetemcomitans cytolethal distending toxin (Cdt) in that cell cycle arrest (G2/M) occurs without leading to apoptosis. We now demonstrate that Cdt-induced cell cycle arrest in OE has a duration of at least 7 days with no change in viability. Moreover, toxin-treated OE develops a new phenotype consistent with cellular senescence; this includes increased senescence-associated β-galactosidase (SA-β-gal) activity and accumulation of the lipopigment, lipofuscin. Moreover, the cells exhibit a secretory profile associated with cellular senescence known as the senescence-associated secretory phenotype (SASP), which includes IL-6, IL-8 and RANKL. Another unique feature of Cdt-induced OE senescence is disruption of barrier function, as shown by loss of transepithelial electrical resistance and confocal microscopic assessment of primary gingival keratinocyte structure. Finally, we demonstrate that Cdt-induced senescence is dependent upon the host cell protein cellugyrin, a homologue of the synaptic vesicle protein synaptogyrin. Collectively, these observations point to a novel pathogenic outcome in oral epithelium that we propose contributes to both A. actinomycetemcomitans infection and periodontal disease progression.
Collapse
Affiliation(s)
- Bruce J. Shenker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Jonathan Korostoff
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (T.J.K.)
| | - Lisa P. Walker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Ali Zekavat
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Anuradha Dhingra
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Taewan J. Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (T.J.K.)
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| |
Collapse
|
4
|
Host Chromatin Regulators Required for Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin Activity in Saccharomyces cerevisiae Model. Infect Immun 2021; 89:e0003621. [PMID: 33941581 DOI: 10.1128/iai.00036-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytolethal distending toxin (CDT) is a bacterial genotoxin that causes host cell cycle arrest and death. We previously employed a Saccharomyces cerevisiae model with inducible expression of the CDT catalytic subunit from Aggregatibacter actinomycetemcomitans, AaCdtB, and showed that a wide variety of host factors play a role in facilitating the activity of CdtB. Our observation that a yeast H2B mutant defective in chromatin condensation was partially resistant to CdtB implies that chromatin structure may affect CDT function. In this study, we identified host chromatin regulatory genes required for CdtB cytotoxicity. We found that the deletion of HTZ1 or certain subunits of SWR, INO80, and SIR complexes increased cellular resistance to CdtB. We hypothesized that CdtB may interact with Htz1 or the chromatin, but immunoprecipitation experiments failed to detect physical interaction between CdtB and Htz1 or the chromatin. However, we observed reduced nuclear localization of CdtB in several mutants, suggesting that impaired nuclear translocation may, at least partly, explain the mechanisms of CdtB resistance. In addition, mutations in chromatin regulatory genes induce changes in the global gene expression profile, and these may indirectly affect CdtB toxicity. Our results suggest that decreased expression of endoplasmic reticulum (ER)-Golgi transport-related genes that may be involved in CdtB transport and/or increased expression of DNA repair genes may contribute to CdtB resistance. These results suggest that the functions of chromatin regulators may contribute to the activity of CDT in host cells.
Collapse
|
5
|
Bharathi V, Girdhar A, Patel BK. Role of CNC1 gene in TDP-43 aggregation-induced oxidative stress-mediated cell death in S. cerevisiae model of ALS. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118993. [PMID: 33647321 DOI: 10.1016/j.bbamcr.2021.118993] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 02/15/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
TDP-43 protein is found deposited as inclusions in the amyotrophic lateral sclerosis (ALS) patient's brain. The mechanism of neuron death in ALS is not fully deciphered but several TDP-43 toxicity mechanisms such as mis-regulation of autophagy, mitochondrial impairment and generation of oxidative stress etc., have been implicated. A predominantly nuclear protein, Cyclin C, can regulate the oxidative stress response via transcription of stress response genes and also by translocation to the cytoplasm for the activation of mitochondrial fragmentation-dependent cell death pathway. Using the well-established yeast TDP-43 proteinopathy model, we examined here whether upon TDP-43 aggregation, cell survival depends on the CNC1 gene that encodes the Cyclin C protein or other genes which encode proteins that function in conjunction with Cyclin C, such as DNM1, FIS1 and MED13. We show that the TDP-43's toxicity is significantly reduced in yeast deleted for CNC1 or DNM1 genes and remains unaltered by deletions of genes, FIS1 and MED13. Importantly, this rescue is observed only in presence of functional mitochondria. Also, deletion of the YBH3 gene involved in the mitochondria-dependent apoptosis pathway reduced the TDP-43 toxicity. Deletion of the VPS1 gene involved in the peroxisomal fission pathway did not mitigate the TDP-43 toxicity. Strikingly, Cyclin C-YFP was observed to relocate to the cytoplasm in response to TDP-43's co-expression which was prevented by addition of an anti-oxidant molecule, N-acetyl cysteine. Overall, the Cyclin C, Dnm1 and Ybh3 proteins are found to be important players in the TDP-43-induced oxidative stress-mediated cell death in the S. cerevisiae model.
Collapse
Affiliation(s)
- Vidhya Bharathi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502285, India
| | - Amandeep Girdhar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502285, India
| | - Basant K Patel
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502285, India.
| |
Collapse
|
6
|
Cytolethal Distending Toxin Subunit B: A Review of Structure-Function Relationship. Toxins (Basel) 2019; 11:toxins11100595. [PMID: 31614800 PMCID: PMC6832162 DOI: 10.3390/toxins11100595] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 01/27/2023] Open
Abstract
The Cytolethal Distending Toxin (CDT) is a bacterial virulence factor produced by several Gram-negative pathogenic bacteria. These bacteria, found in distinct niches, cause diverse infectious diseases and produce CDTs differing in sequence and structure. CDTs have been involved in the pathogenicity of the associated bacteria by promoting persistent infection. At the host-cell level, CDTs cause cell distension, cell cycle block and DNA damage, eventually leading to cell death. All these effects are attributable to the catalytic CdtB subunit, but its exact mode of action is only beginning to be unraveled. Sequence and 3D structure analyses revealed similarities with better characterized proteins, such as nucleases or phosphatases, and it has been hypothesized that CdtB exerts a biochemical activity close to those enzymes. Here, we review the relationships that have been established between CdtB structure and function, particularly by mutation experiments on predicted key residues in different experimental systems. We discuss the relevance of these approaches and underline the importance of further study in the molecular mechanisms of CDT toxicity, particularly in the context of different pathological conditions.
Collapse
|
7
|
Denmongkholchai S, Katare P, Choochuay S, Thanyasrisung P, Tsuruda K, Sugai M, Mongkolsuk S, Matangkasombut O. Genome-Wide Identification of Host Genes Required for Toxicity of Bacterial Cytolethal Distending Toxin in a Yeast Model. Front Microbiol 2019; 10:890. [PMID: 31080443 PMCID: PMC6497811 DOI: 10.3389/fmicb.2019.00890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/08/2019] [Indexed: 12/16/2022] Open
Abstract
Background Aggregatibacter actinomycetemcomitans, a periodontal pathogen, secretes a cytolethal distending toxin (AaCDT) that causes host cell cycle arrest and cell death. Although CDT could be an important virulence factor, it is unclear how it enters the nucleus to exert its cytotoxicity. Objective To investigate the mechanisms of AaCDT by genome-wide screening for host mutations that confer resistance to the catalytic subunit, AaCdtB, in a Saccharomyces cerevisiae model. Methods We transformed the yeast haploid deletion library, a collection of yeast mutants with single gene deletions of virtually all non-essential ORFs in the genome, with plasmids carrying galactose-inducible AaCdtB. Yeast mutants that showed resistance to AaCdtB were selected and rescreened by a spotting assay. AaCdtB expression was confirmed by western blot analysis; any mutants that showed no or weak expression of AaCdtB were omitted from the analysis. The lists of genes whose mutations confer resistance to AaCdtB were analyzed for Gene Ontology (GO) term enrichments. Localization of AaCdtB-EGFP was examined using fluorescent microscopy. Nuclear localization relative to EGFP control was calculated and compared to wild-type. Results Out of approximately 5,000 deletion mutants, we isolated 243 mutants that are resistant to AaCdtB. GO analyses indicated that genes associated with organic anion transport are significantly enriched (16 genes). Furthermore, several genes associated with the nucleus and endoplasmic reticulum (ER) were identified. Localization studies of AaCdtB, in mutants with the deletion of genes associated with the GO term organic anion transport, showed lower nuclear localization than wild-type. The results suggest that these genes may be required for AaCdtB translocation into the nucleus and its cytotoxicity. Conclusion The genome-wide screen in the yeast deletion library allowed us to identify a large number of host genes required for AaCdtB cytotoxicity. Further investigation could lead to more insights into the mechanisms of CdtB intoxication.
Collapse
Affiliation(s)
- Siriyod Denmongkholchai
- Interdepartmental Program in Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand.,Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prashant Katare
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Graduate Program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sarocha Choochuay
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Panida Thanyasrisung
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Keiko Tsuruda
- Department of Oral Epidemiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Motoyuki Sugai
- Department of Antimicrobial Resistance, Graduate School of Biomedical and Health Sciences, Project Research Center for Nosocomial Infectious Diseases (RCNID), Hiroshima University, Hiroshima, Japan.,Antimicrobial Resistance Research Center, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Skorn Mongkolsuk
- Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Oranart Matangkasombut
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Laboratory of Biotechnology, Chulabhorn Research Institute, Bangkok, Thailand
| |
Collapse
|
8
|
CdtC-Induced Processing of Membrane-Bound CdtA Is a Crucial Step in Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin Holotoxin Formation. Infect Immun 2018; 86:IAI.00731-17. [PMID: 29229729 DOI: 10.1128/iai.00731-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/02/2017] [Indexed: 11/20/2022] Open
Abstract
Aggregatibacter actinomycetemcomitans is an oral pathogen causing periodontal disease and bacterial endocarditis. It produces cytolethal distending toxin (CDT) that could damage mammalian cells and tissues. CDT is a tripartite protein toxin composed of CdtA, CdtB, and CdtC. We have previously indicated that CdtA is a lipoprotein and that the proteolytic processing of CdtA is important for biogenesis and secretion of CDT holotoxin. Here, we established an in vitro processing assay of CdtA and investigated the interactions of CdtA with other Cdt subunits. This assay demonstrated that incubation of membrane-bound CdtA (MCdtA), CdtB, and CdtC immediately generated a processed form of CdtA (CdtA'), which is recovered from the soluble fraction. In contrast, incubation of soluble membrane-unbound CdtA with CdtB and CdtC did not yield any CdtA'. Furthermore, incubation of CdtC with MCdtA was enough to induce rapid processing of MCdtA, whereas CdtB alone was unable to induce the processing. Coimmunoprecipitation demonstrated that CdtA' and CdtC formed a complex. Furthermore, subsequent addition of CdtB to this reaction mixture resulted in complete CDT holotoxin complex. The cytolethal distending activity assay demonstrated that CDT complex containing CdtA' showed far stronger cytotoxicity than that containing CdtA. Collectively, our data suggest that CDT holotoxin formation in vivo is a sequential event: interaction of MCdtA and CdtC induces proteolytic processing of MCdtA, and the released CdtA' forms a complex with CdtC. Subsequent binding of CdtB to the CdtA'/CdtC complex results in CDT holotoxin formation.
Collapse
|
9
|
Scuron MD, Boesze-Battaglia K, Dlakić M, Shenker BJ. The Cytolethal Distending Toxin Contributes to Microbial Virulence and Disease Pathogenesis by Acting As a Tri-Perditious Toxin. Front Cell Infect Microbiol 2016; 6:168. [PMID: 27995094 PMCID: PMC5136569 DOI: 10.3389/fcimb.2016.00168] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/15/2016] [Indexed: 12/11/2022] Open
Abstract
This review summarizes the current status and recent advances in our understanding of the role that the cytolethal distending toxin (Cdt) plays as a virulence factor in promoting disease by toxin-producing pathogens. A major focus of this review is on the relationship between structure and function of the individual subunits that comprise the AB2 Cdt holotoxin. In particular, we concentrate on the molecular mechanisms that characterize this toxin and which account for the ability of Cdt to intoxicate multiple cell types by utilizing a ubiquitous binding partner on the cell membrane. Furthermore, we propose a paradigm shift for the molecular mode of action by which the active Cdt subunit, CdtB, is able to block a key signaling cascade and thereby lead to outcomes based upon programming and the role of the phosphatidylinositol 3-kinase (PI-3K) in a variety of cells. Based upon the collective Cdt literature, we now propose that Cdt is a unique and potent virulence factor capable of acting as a tri-perditious toxin that impairs host defenses by: (1) disrupting epithelial barriers; (2) suppressing acquired immunity; (3) promoting pro-inflammatory responses. Thus, Cdt plays a key role in facilitating the early stages of infection and the later stages of disease progression by contributing to persistence and impairing host elimination.
Collapse
Affiliation(s)
- Monika D Scuron
- Department of Pathology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Mensur Dlakić
- Department of Microbiology and Immunology, Montana State University Bozeman, MT, USA
| | - Bruce J Shenker
- Department of Pathology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
10
|
Cell resistance to the Cytolethal Distending Toxin involves an association of DNA repair mechanisms. Sci Rep 2016; 6:36022. [PMID: 27775089 PMCID: PMC5075911 DOI: 10.1038/srep36022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/10/2016] [Indexed: 02/06/2023] Open
Abstract
The Cytolethal Distending Toxin (CDT), produced by many bacteria, has been associated with various diseases including cancer. CDT induces DNA double-strand breaks (DSBs), leading to cell death or mutagenesis if misrepaired. At low doses of CDT, other DNA lesions precede replication-dependent DSB formation, implying that non-DSB repair mechanisms may contribute to CDT cell resistance. To address this question, we developed a proliferation assay using human cell lines specifically depleted in each of the main DNA repair pathways. Here, we validate the involvement of the two major DSB repair mechanisms, Homologous Recombination and Non Homologous End Joining, in the management of CDT-induced lesions. We show that impairment of single-strand break repair (SSBR), but not nucleotide excision repair, sensitizes cells to CDT, and we explore the interplay of SSBR with the DSB repair mechanisms. Finally, we document the role of the replicative stress response and demonstrate the involvement of the Fanconi Anemia repair pathway in response to CDT. In conclusion, our work indicates that cellular survival to CDT-induced DNA damage involves different repair pathways, in particular SSBR. This reinforces a model where CDT-related genotoxicity primarily involves SSBs rather than DSBs, underlining the importance of cell proliferation during CDT intoxication and pathogenicity.
Collapse
|
11
|
Jaffar N, Miyazaki T, Maeda T. Biofilm formation of periodontal pathogens on hydroxyapatite surfaces: Implications for periodontium damage. J Biomed Mater Res A 2016; 104:2873-80. [PMID: 27390886 DOI: 10.1002/jbm.a.35827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/24/2016] [Accepted: 07/06/2016] [Indexed: 11/05/2022]
Abstract
Biofilm formation of periodontal pathogens on teeth surfaces promotes the progression of periodontal disease. Hence, understanding the mechanisms of bacterial attachment to the dental surfaces may inform strategies for the maintenance of oral health. Although hydroxyapatite (HA) is a major calcium phosphate component of teeth, effect of biofilm formation on HA surfaces remains poorly characterized. In this study, biofilm-forming abilities by the periodontal pathogens Aggregatibacter actinomycetemcomitans Y4 and Porphyromonas gingivalis 381 were investigated on dense and porous HAs that represent enamel and dentin surfaces, respectively. These experiments showed greater biofilm formation on porous HA, but differing attachment profiles and effects of the two pathogens. Specifically, while the detachment of A. actinomycetemcomitans Y4 biofilm was observed, P. gingivalis 381 biofilm increased with time. Moreover, observations of HA morphology following formation of A. actinomycetemcomitans Y4 biofilm revealed gaps between particles, whereas no significant changes were observed in the presence of P. gingivalis 381. Finally, comparisons of calcium leakage showed only slight differences between bacterial species and HA types and may be masked by bacterial calcium uptake. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2873-2880, 2016.
Collapse
Affiliation(s)
- Norzawani Jaffar
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0196, Japan.,Faculty of Health Sciences, Gong Badak Campus, Universiti Sultan Zainal Abidin (UniSZA), Kuala Terengganu, Terengganu Darul Iman, Malaysia
| | - Toshiki Miyazaki
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0196, Japan
| | - Toshinari Maeda
- Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, 2-4 Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0196, Japan. .,Research Center for Advanced Eco-Fitting Technology, Kyushu Institute of Technology, Kitakyushu, Japan.
| |
Collapse
|
12
|
Jaffar N, Ishikawa Y, Mizuno K, Okinaga T, Maeda T. Mature Biofilm Degradation by Potential Probiotics: Aggregatibacter actinomycetemcomitans versus Lactobacillus spp. PLoS One 2016; 11:e0159466. [PMID: 27438340 PMCID: PMC4954673 DOI: 10.1371/journal.pone.0159466] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 07/01/2016] [Indexed: 02/07/2023] Open
Abstract
The biofilm degradation of Aggregatibacter actinomycetemcomitans is essential as a complete periodontal disease therapy, and here we show the effects of potential probiotic bacteria such as Lactobacillus spp. for the biofilm of several serotypes of A. actinomycetemcomitans strains. Eight of the 13 species showed the competent biofilm degradation of ≥ 90% reduction in biofilm values in A. actinomycetemcomitans Y4 (serotype b) as well as four of the seven species for the biofilm of A. actinomycetemcomitans OMZ 534 (serotype e). In contrast, the probiotic bacteria did not have a big impact for the degradation of A. actinomycetemcomitans SUNY 75 (serotype a) biofilm. The dispersed A. actinomycetemcomitans Y4 cells through the biofilm detachment were still viable and plausible factors for the biofilm degradation were not due to the lactic acid and low pH conditions. The three enzymes, protease, lipase, and amylase may be responsible for the biofilm degradation; in particular, lipase was the most effective enzyme for the biofilm degradation of A. actinomycetemcomitans Y4 along with the protease activity which should be also important for the other serotypes. Remarkable lipase enzyme activities were detected from some of the potential probiotics and a supporting result using a lipase inhibitor presented corroborating evidence that lipase activity is one of the contributing factors for biofilm degradation outside of the protease which is also another possible factor for the biofilm of the other serotype of A. actinomycetemcomitans strains. On the other hand, the biofilm of A. actinomycetemcomitans SUNY 75 (serotype a) was not powerfully degraded by the lipase enzyme because the lipase inhibitor was slightly functional for only two of potential probiotics.
Collapse
Affiliation(s)
- Norzawani Jaffar
- Department of Biological Functions Engineering, Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, Wakamatsu-ku, Kitakyushu, Japan
- Faculty of Health Sciences, Gong Badak Campus, Universiti Sultan Zainal Abidin (UniSZA), Kuala Terengganu, Terengganu Darul Iman, Malaysia
| | - Yuya Ishikawa
- Department of Biological Functions Engineering, Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, Wakamatsu-ku, Kitakyushu, Japan
| | - Kouhei Mizuno
- Department of Materials Science and Chemical Engineering, Kitakyushu National College of Technology, Kitakyushu, Japan
| | - Toshinori Okinaga
- School of Oral Health Sciences, Faculty of Dentistry, Kyushu Dental University, Kitakyushu, Japan
| | - Toshinari Maeda
- Department of Biological Functions Engineering, Graduate School of Life Science and Systems Engineering, Kyushu Institute of Technology, Wakamatsu-ku, Kitakyushu, Japan
- Research Center for Advanced Eco-fitting Technology, Kyushu Institute of Technology, Kitakyushu, Japan
- * E-mail:
| |
Collapse
|
13
|
Obradović D, Gašperšič R, Caserman S, Leonardi A, Jamnik M, Podlesek Z, Seme K, Anderluh G, Križaj I, Maček P, Butala M. A Cytolethal Distending Toxin Variant from Aggregatibacter actinomycetemcomitans with an Aberrant CdtB That Lacks the Conserved Catalytic Histidine 160. PLoS One 2016; 11:e0159231. [PMID: 27414641 PMCID: PMC4945079 DOI: 10.1371/journal.pone.0159231] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/29/2016] [Indexed: 12/19/2022] Open
Abstract
The periodontopathogen Aggregatibacter actinomycetemcomitans synthesizes several virulence factors, including cytolethal distending toxin (CDT). The active CDT holoenzyme is an AB-type tripartite genotoxin that affects eukaryotic cells. Subunits CdtA and CdtC (B-components) allow binding and intracellular translocation of the active CdtB (A-component), which elicits nuclear DNA damage. Different strains of A. actinomycetemcomitans have diverse virulence genotypes, which results in varied pathogenic potential and disease progression. Here, we identified an A. actinomycetemcomitans strain isolated from two patients with advance chronic periodontitis that has a regular cdtABC operon, which, however, codes for a unique, shorter, variant of the CdtB subunit. We describe the characteristics of this CdtBΔ116–188, which lacks the intact nuclear localisation signal and the catalytic histidine 160. We show that the A. actinomycetemcomitans DO15 isolate secretes CdtBΔ116–188, and that this subunit cannot form a holotoxin and is also not genotoxic if expressed ectopically in HeLa cells. Furthermore, the A. actinomycetemcomitans DO15 isolate is not toxic, nor does it induce cellular distention upon infection of co-cultivated HeLa cells. Biological significance of this deletion in the cdtB remains to be explained.
Collapse
Affiliation(s)
- Davor Obradović
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Rok Gašperšič
- Department of Oral Medicine and Periodontology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Simon Caserman
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Adrijana Leonardi
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Maja Jamnik
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Zdravko Podlesek
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Katja Seme
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Anderluh
- Laboratory for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Igor Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Maček
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
- * E-mail: (PM); (MB)
| | - Matej Butala
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
- * E-mail: (PM); (MB)
| |
Collapse
|
14
|
Shenker BJ, Boesze-Battaglia K, Scuron MD, Walker LP, Zekavat A, Dlakić M. The toxicity of the Aggregatibacter actinomycetemcomitans cytolethal distending toxin correlates with its phosphatidylinositol-3,4,5-triphosphate phosphatase activity. Cell Microbiol 2015; 18:223-43. [PMID: 26247396 DOI: 10.1111/cmi.12497] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/22/2015] [Accepted: 07/26/2015] [Indexed: 12/22/2022]
Abstract
The Aggregatibacter actinomycetemcomitans cytolethal distending toxin (Cdt) induces G2 arrest and apoptosis in lymphocytes and other cell types. We have shown that the active subunit, CdtB, exhibits phosphatidylinositol-3,4,5-triphosphate (PIP3) phosphatase activity, leading us to propose that Cdt toxicity is the result of PIP3 depletion and perturbation of phosphatidylinositol-3-kinase (PI-3K)/PIP3/Akt signalling. To further explore this relationship, we have focused our analysis on identifying residues that comprise the catalytic pocket and are critical to substrate binding rather than catalysis. In this context, we have generated several CdtB mutants and demonstrate that, in each instance, the ability of the toxin to induce cell cycle arrest correlates with retention of phosphatase activity. We have also assessed the effect of Cdt on downstream components of the PI-3K signalling pathway. In addition to depletion of intracellular concentrations of PIP3, toxin-treated lymphocytes exhibit decreases in pAkt and pGSK3β. Further analysis indicates that toxin-treated cells exhibit a concomitant loss in Akt activity and increase in GSK3β kinase activity consistent with observed changes in their phosphorylation status. We demonstrate that cell susceptibility to Cdt is dependent upon dephosphorylation and concomitant activation of GSK3β. Finally, we demonstrate that, in addition to lymphocytes, HeLa cells exposed to a CdtB mutant that retains phosphatase activity and not DNase activity undergo G2 arrest in the absence of H2AX phosphorylation. Our results provide further insight into the mode of action by which Cdt may function as an immunotoxin and induce cell cycle arrest in target cells such as lymphocytes.
Collapse
Affiliation(s)
- Bruce J Shenker
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Monika Damek Scuron
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Lisa P Walker
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Ali Zekavat
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Mensur Dlakić
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| |
Collapse
|
15
|
Khairalla AS, Omer SA, Mahdavi J, Aslam A, Dufailu OA, Self T, Jonsson AB, Geörg M, Sjölinder H, Royer PJ, Martinez-Pomares L, Ghaemmaghami AM, Wooldridge KG, Oldfield NJ, Ala'Aldeen DAA. Nuclear trafficking, histone cleavage and induction of apoptosis by the meningococcal App and MspA autotransporters. Cell Microbiol 2015; 17:1008-20. [PMID: 25600171 PMCID: PMC5024080 DOI: 10.1111/cmi.12417] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 12/18/2014] [Accepted: 01/13/2015] [Indexed: 01/13/2023]
Abstract
Neisseria meningitidis, a major cause of bacterial meningitis and septicaemia, secretes multiple virulence factors, including the adhesion and penetration protein (App) and meningococcal serine protease A (MspA). Both are conserved, immunogenic, type Va autotransporters harbouring S6‐family serine endopeptidase domains. Previous work suggested that both could mediate adherence to human cells, but their precise contribution to meningococcal pathogenesis was unclear. Here, we confirm that App and MspA are in vivo virulence factors since human CD46‐expressing transgenic mice infected with meningococcal mutants lacking App, MspA or both had improved survival rates compared with mice infected with wild type. Confocal imaging showed that App and MspA were internalized by human cells and trafficked to the nucleus. Cross‐linking and enzyme‐linked immuno assay (ELISA) confirmed that mannose receptor (MR), transferrin receptor 1 (TfR1) and histones interact with MspA and App. Dendritic cell (DC) uptake could be blocked using mannan and transferrin, the specific physiological ligands for MR and TfR1, whereas in vitro clipping assays confirmed the ability of both proteins to proteolytically cleave the core histone H3. Finally, we show that App and MspA induce a dose‐dependent increase in DC death via caspase‐dependent apoptosis. Our data provide novel insights into the roles of App and MspA in meningococcal infection.
Collapse
Affiliation(s)
| | - Sherko A Omer
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Jafar Mahdavi
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Akhmed Aslam
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Osman A Dufailu
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Tim Self
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Ann-Beth Jonsson
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Miriam Geörg
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Hong Sjölinder
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | | | | | | | - Neil J Oldfield
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
16
|
Bachtiar EW, Bachtiar BM, Jarosz LM, Amir LR, Sunarto H, Ganin H, Meijler MM, Krom BP. AI-2 of Aggregatibacter actinomycetemcomitans inhibits Candida albicans biofilm formation. Front Cell Infect Microbiol 2014; 4:94. [PMID: 25101248 PMCID: PMC4104835 DOI: 10.3389/fcimb.2014.00094] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/26/2014] [Indexed: 12/14/2022] Open
Abstract
Aggregatibacter actinomycetemcomitans, a Gram-negative bacterium, and Candida albicans, a polymorphic fungus, are both commensals of the oral cavity but both are opportunistic pathogens that can cause oral diseases. A. actinomycetemcomitans produces a quorum-sensing molecule called autoinducer-2 (AI-2), synthesized by LuxS, that plays an important role in expression of virulence factors, in intra- but also in interspecies communication. The aim of this study was to investigate the role of AI-2 based signaling in the interactions between C. albicans and A. actinomycetemcomitans. A. actinomycetemcomitans adhered to C. albicans and inhibited biofilm formation by means of a molecule that was secreted during growth. C. albicans biofilm formation increased significantly when co-cultured with A. actinomycetemcomitans luxS, lacking AI-2 production. Addition of wild-type-derived spent medium or synthetic AI-2 to spent medium of the luxS strain, restored inhibition of C. albicans biofilm formation to wild-type levels. Addition of synthetic AI-2 significantly inhibited hypha formation of C. albicans possibly explaining the inhibition of biofilm formation. AI-2 of A. actinomycetemcomitans is synthesized by LuxS, accumulates during growth and inhibits C. albicans hypha- and biofilm formation. Identifying the molecular mechanisms underlying the interaction between bacteria and fungi may provide important insight into the balance within complex oral microbial communities.
Collapse
Affiliation(s)
- Endang W Bachtiar
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia Jakarta, Indonesia
| | - Boy M Bachtiar
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia Jakarta, Indonesia
| | - Lucja M Jarosz
- Department of Biomedical Engineering, The W.J. Kolff Institute, University Medical Center Groningen and University of Groningen Groningen, Netherlands
| | - Lisa R Amir
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia Jakarta, Indonesia
| | - Hari Sunarto
- Department of Periodontology, Faculty of Dentistry, Universitas Indonesia Jakarta, Indonesia
| | - Hadas Ganin
- Department of Chemistry, Ben-Gurion University of the Negev Be'er-Sheva, Israel
| | - Michael M Meijler
- Department of Chemistry, Ben-Gurion University of the Negev Be'er-Sheva, Israel
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Free University Amsterdam Amsterdam, Netherlands
| |
Collapse
|
17
|
Bezine E, Vignard J, Mirey G. The cytolethal distending toxin effects on Mammalian cells: a DNA damage perspective. Cells 2014; 3:592-615. [PMID: 24921185 PMCID: PMC4092857 DOI: 10.3390/cells3020592] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 12/27/2022] Open
Abstract
The cytolethal distending toxin (CDT) is produced by many pathogenic Gram-negative bacteria and is considered as a virulence factor. In human cells, CDT exposure leads to a unique cytotoxicity associated with a characteristic cell distension and induces a cell cycle arrest dependent on the DNA damage response (DDR) triggered by DNA double-strand breaks (DSBs). CDT has thus been classified as a cyclomodulin and a genotoxin. Whereas unrepaired damage can lead to cell death, effective, but improper repair may be detrimental. Indeed, improper repair of DNA damage may allow cells to resume the cell cycle and induce genetic instability, a hallmark in cancer. In vivo, CDT has been shown to induce the development of dysplastic nodules and to lead to genetic instability, defining CDT as a potential carcinogen. It is therefore important to characterize the outcome of the CDT-induced DNA damage and the consequences for intoxicated cells and organisms. Here, we review the latest results regarding the host cell response to CDT intoxication and focus on DNA damage characteristics, cell cycle modulation and cell outcomes.
Collapse
Affiliation(s)
- Elisabeth Bezine
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France.
| | - Julien Vignard
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France.
| | - Gladys Mirey
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France.
| |
Collapse
|
18
|
Probing bacterial metabolism during infection using high-resolution transcriptomics. J Bacteriol 2013; 195:4991-8. [PMID: 23974023 DOI: 10.1128/jb.00875-13] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A fundamental aspect of most infectious diseases is the need for the invading microbe to proliferate in the host. However, little is known about the metabolic pathways required for pathogenic microbes to colonize and persist in their hosts. In this study, we used RNA sequencing (RNA-seq) to generate a high-resolution transcriptome of the opportunistic pathogen Aggregatibacter actinomycetemcomitans in vivo. We identified 691 A. actinomycetemcomitans transcriptional start sites and 210 noncoding RNAs during growth in vivo and as a biofilm in vitro. Compared to in vitro biofilm growth on a defined medium, ∼14% of the A. actinomycetemcomitans genes were differentially regulated in vivo. A disproportionate number of genes coding for proteins involved in metabolic pathways were differentially regulated in vivo, suggesting that A. actinomycetemcomitans in vivo metabolism is distinct from in vitro growth. Mutational analyses of differentially regulated genes revealed that formate dehydrogenase H and fumarate reductase are important A. actinomycetemcomitans fitness determinants in vivo. These results not only provide a high-resolution genomic analysis of a bacterial pathogen during in vivo growth but also provide new insight into metabolic pathways required for A. actinomycetemcomitans in vivo fitness.
Collapse
|
19
|
Prediction and screening of nuclear targeting proteins with nuclear localization signals in Helicobacter pylori. J Microbiol Methods 2012; 91:490-6. [PMID: 23079023 DOI: 10.1016/j.mimet.2012.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/05/2012] [Accepted: 10/09/2012] [Indexed: 01/26/2023]
Abstract
Host cell pathology induced by nuclear targeting of bacterial proteins has recently been identified as a pathogenic mechanism of bacteria. However, very few bacterial proteins were identified to target the nuclei of host cells. This study was designed to screen nuclear targeting proteins with nuclear localization signals (NLSs) in Helicobacter pylori using a combination of bioinformatic analysis and the Gateway recombinational cloning system. Forty-nine functional or hypothetical proteins were predicted to carry the putative NLSs among 1570 open reading frames (ORFs) of H. pylori 26695. Entire sets of 49 H. pylori ORFs were cloned for the generation of green fluorescent protein-tagged proteins using the Gateway recombinational cloning system. Twenty-six H. pylori proteins with the putative NLSs were found to target in the nuclei of COS-7 cells, whereas 23 were localized in the cytoplasm of host cells. Deletion of NLS sequences from four selected nuclear targeting proteins, urease subunit A, Omp18, secreted protein involved in flagellar motility, and response regulator, resulted in cytoplasmic localization of these mutant proteins. In conclusion, a combination of bioinformatic analysis and the Gateway cloning system was shown to be a useful tool for large-scale screening of nuclear targeting proteins with NLSs in H. pylori, which can be used to better understand the H. pylori-directed host cell pathology.
Collapse
|
20
|
Fedor Y, Vignard J, Nicolau-Travers ML, Boutet-Robinet E, Watrin C, Salles B, Mirey G. From single-strand breaks to double-strand breaks during S-phase: a new mode of action of theEscherichia coli Cytolethal Distending Toxin. Cell Microbiol 2012; 15:1-15. [DOI: 10.1111/cmi.12028] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 12/12/2022]
Affiliation(s)
- Y. Fedor
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - J. Vignard
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - M.-L. Nicolau-Travers
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - E. Boutet-Robinet
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - C. Watrin
- INRA; USC 1360; F-31300 Toulouse France
- Université de Toulouse; UPS; Centre de Physiopathologie de Toulouse Purpan (CPTP); F-31400 Toulouse France
- Inserm; UMR1043; F-31300 Toulouse France
- CNRS; UMR5282; F-31400 Toulouse France
| | - B. Salles
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - G. Mirey
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| |
Collapse
|
21
|
Gargi A, Reno M, Blanke SR. Bacterial toxin modulation of the eukaryotic cell cycle: are all cytolethal distending toxins created equally? Front Cell Infect Microbiol 2012; 2:124. [PMID: 23061054 PMCID: PMC3465861 DOI: 10.3389/fcimb.2012.00124] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Accepted: 09/17/2012] [Indexed: 01/08/2023] Open
Abstract
The cytolethal distending toxins (CDTs) comprise a family of intracellular-acting bacterial protein toxins whose actions upon eukaryotic cells result in several consequences, the most characteristic of which is the induction of G(2)/M cell cycle arrest. Most CDTs are hetero-tripartite assemblies of CdtA, CdtB, and CdtC, with CdtB required for CDT-mediated cell cycle arrest. Several lines of evidence indicate that CdtA and CdtC are required for the optimal intracellular activity of CdtB, although the exact functional roles of CdtA and CdtC remain poorly understood. The genes encoding the CDTs have been identified in a diverse array of Gram-negative pathogenic bacteria. More recently, the genes encoding several CdtB subunits have been associated with alternatively linked subunits resembling the B-subunits of pertussis toxin. Although the CDTs are generally considered to all function as bacterial genotoxins, the extent to which individual members of the CDTs employ similar mechanisms of cell surface binding, uptake, and trafficking within sensitive cells is poorly understood. Recently, data have begun to emerge suggesting differences in the molecular basis by which individual CDTs interact with and enter host cells, suggesting the possibility that CDTs possess properties reflecting the specific niches idiosyncratic to those CDT bacterial pathogens that produce them. The extent to which functional differences between individual CDTs reflect the specific requirements for intoxicating cells and tissues within the diverse range of host microenvironments colonized by CDT-producing pathogenic bacteria remains to be experimentally explored.
Collapse
Affiliation(s)
| | | | - Steven R. Blanke
- Department of Microbiology, Institute for Genomic Biology, University of IllinoisUrbana, IL, USA
| |
Collapse
|
22
|
Guerra L, Guidi R, Frisan T. Do bacterial genotoxins contribute to chronic inflammation, genomic instability and tumor progression? FEBS J 2011; 278:4577-88. [DOI: 10.1111/j.1742-4658.2011.08125.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
23
|
Jinadasa RN, Bloom SE, Weiss RS, Duhamel GE. Cytolethal distending toxin: a conserved bacterial genotoxin that blocks cell cycle progression, leading to apoptosis of a broad range of mammalian cell lineages. MICROBIOLOGY-SGM 2011; 157:1851-1875. [PMID: 21565933 DOI: 10.1099/mic.0.049536-0] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cytolethal distending toxin (CDT) is a heterotrimeric AB-type genotoxin produced by several clinically important Gram-negative mucocutaneous bacterial pathogens. Irrespective of the bacterial species of origin, CDT causes characteristic and irreversible cell cycle arrest and apoptosis in a broad range of cultured mammalian cell lineages. The active subunit CdtB has structural homology with the phosphodiesterase family of enzymes including mammalian DNase I, and alone is necessary and sufficient to account for cellular toxicity. Indeed, mammalian cells treated with CDT initiate a DNA damage response similar to that elicited by ionizing radiation-induced DNA double strand breaks resulting in cell cycle arrest and apoptosis. The mechanism of CDT-induced apoptosis remains incompletely understood, but appears to involve both p53-dependent and -independent pathways. While epithelial, endothelial and fibroblast cell lines respond to CDT by undergoing arrest of cell cycle progression resulting in nuclear and cytoplasmic distension that precedes apoptotic cell death, cells of haematopoietic origin display rapid apoptosis following a brief period of cell cycle arrest. In this review, the ecology of pathogens producing CDT, the molecular biology of bacterial CDT and the molecular mechanisms of CDT-induced cytotoxicity are critically appraised. Understanding the contribution of a broadly conserved bacterial genotoxin that blocks progression of the mammalian cell cycle, ultimately causing cell death, should assist with elucidating disease mechanisms for these important pathogens.
Collapse
Affiliation(s)
- Rasika N Jinadasa
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Stephen E Bloom
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Robert S Weiss
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Gerald E Duhamel
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
24
|
Genetic constructions, hyperexpressing recombinant fragments of cytolethal distending toxin of Aggregatibacter actinomycetemcomitans. World J Microbiol Biotechnol 2011. [DOI: 10.1007/s11274-010-0567-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Guerra L, Cortes-Bratti X, Guidi R, Frisan T. The biology of the cytolethal distending toxins. Toxins (Basel) 2011; 3:172-90. [PMID: 22069704 PMCID: PMC3202825 DOI: 10.3390/toxins3030172] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 02/14/2011] [Accepted: 02/22/2011] [Indexed: 12/11/2022] Open
Abstract
The cytolethal distending toxins (CDTs), produced by a variety of Gram-negative pathogenic bacteria, are the first bacterial genotoxins described, since they cause DNA damage in the target cells. CDT is an A-B(2) toxin, where the CdtA and CdtC subunits are required to mediate the binding on the surface of the target cells, allowing internalization of the active CdtB subunit, which is functionally homologous to the mammalian deoxyribonuclease I. The nature of the surface receptor is still poorly characterized, however binding of CDT requires intact lipid rafts, and its internalization occurs via dynamin-dependent endocytosis. The toxin is retrograde transported through the Golgi complex and the endoplasmic reticulum, and subsequently translocated into the nuclear compartment, where it exerts the toxic activity. Cellular intoxication induces DNA damage and activation of the DNA damage responses, which results in arrest of the target cells in the G1 and/or G2 phases of the cell cycle and activation of DNA repair mechanisms. Cells that fail to repair the damage will senesce or undergo apoptosis. This review will focus on the well-characterized aspects of the CDT biology and discuss the questions that still remain unanswered.
Collapse
Affiliation(s)
- Lina Guerra
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden, Box 285, S-171 77 Stockholm, Sweden.
| | | | | | | |
Collapse
|
26
|
Current awareness on yeast. Yeast 2010. [DOI: 10.1002/yea.1719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|