Bacterial Genotoxin-Induced DNA Damage and Modulation of the Host Immune Microenvironment
Abstract
1. Bacterial Genotoxins
1.1. Cytolethal Distending Toxins And Typhoid Toxin Structure
1.2. CDTs Activity
1.3. Colibactin
2. The DNA Damage Response
3. Modulation of Immune Response by the BTGXs-induced DNA Damage
3.1. The Fine-Tuning of the Immune Response
3.2. BTGXs-Induced DNA Damage and the Immune Response
3.3. Pro-Inflammatory Effects of BTGX
3.4. Immunosuppressive and Anti-Inflammatory Effects of BTGXs
4. Concluding Remarks
Funding
Conflicts of Interest
References
- Frisan, T.; Guidi, R.; Guerra, L. Toxins acting on intracellular targets: Only foes or also friends? In Bacterial Pathogenesis: Molecular and Cellular Mechanisms; Locht, C., Simonet, S., Eds.; Caister Academic Press: Norfolk, UK, 2012. [Google Scholar]
- Grasso, F.; Frisan, T. Bacterial genotoxins: Merging the DNA damage response into infection biology. Biomolecules 2015, 5, 1762–1782. [Google Scholar] [CrossRef]
- Cortes-Bratti, X.; Frisan, T.; Thelestam, M. The cytolethal distending toxins induce DNA damage and cell cycle arrest. Toxicon Off. J. Int. Soc. Toxinol. 2001, 39, 1729–1736. [Google Scholar] [CrossRef]
- Jinadasa, R.N.; Bloom, S.E.; Weiss, R.S.; Duhamel, G.E. Cytolethal distending toxin: A conserved bacterial genotoxin that blocks cell cycle progression, leading to apoptosis of a broad range of mammalian cell lineages. Microbiology 2011, 157 Pt 7, 1851–1875. [Google Scholar] [CrossRef]
- Fais, T.; Delmas, J.; Barnich, N.; Bonnet, R.; Dalmasso, G. Colibactin: More than a new bacterial toxin. Toxins 2018, 10, 151. [Google Scholar] [CrossRef] [PubMed]
- Chumduri, C.; Gurumurthy, R.K.; Zietlow, R.; Meyer, T.F. Subversion of host genome integrity by bacterial pathogens. Nat. Rev. Mol. Cell Biol. 2016, 17, 659–673. [Google Scholar] [CrossRef] [PubMed]
- Nesic, D.; Hsu, Y.; Stebbins, C.E. Assembly and function of a bacterial genotoxin. Nature 2004, 429, 429–433. [Google Scholar] [CrossRef] [PubMed]
- Frisan, T. Bacterial genotoxins. In The Comprehensive Sourcebook of Bacterial Protein Toxins; Alouf, J., Ladant, D., Popoff, M., Eds.; Elsevier: Amsterdam, The Netherlands, 2015; pp. 558–601. [Google Scholar]
- Nesic, D.; Stebbins, C.E. Mechanisms of assembly and cellular interactions for the bacterial genotoxin CDT. PLoS Pathog. 2005, 1, e28. [Google Scholar] [CrossRef] [PubMed]
- Song, J.; Gao, X.; Galan, J.E. Structure and function of the Salmonella Typhi chimaeric A(2)B(5) typhoid toxin. Nature 2013, 499, 350–354. [Google Scholar] [CrossRef] [PubMed]
- Desai, P.T.; Porwollik, S.; Long, F.; Cheng, P.; Wollam, A.; Clifton, S.W.; Weinstock, G.M.; McClelland, M. Evolutionary genomics of Salmonella enterica subspecies. mBio 2013, 4, e00579-12. [Google Scholar] [CrossRef] [PubMed]
- Suez, J.; Porwollik, S.; Dagan, A.; Marzel, A.; Schorr, Y.I.; Desai, P.T.; Agmon, V.; McClelland, M.; Rahav, G.; Gal-Mor, O. Virulence gene profiling and pathogenicity characterization of non-typhoidal salmonella accounted for invasive disease in humans. PLoS ONE 2013, 8, e58449. [Google Scholar] [CrossRef]
- Rodriguez-Rivera, L.D.; Bowen, B.M.; den Bakker, H.C.; Duhamel, G.E.; Wiedmann, M. Characterization of the cytolethal distending toxin (typhoid toxin) in non-typhoidal Salmonella serovars. Gut Pathog. 2015, 7, 19. [Google Scholar] [CrossRef] [PubMed]
- Spano, S.; Ugalde, J.E.; Galan, J.E. Delivery of a Salmonella Typhi exotoxin from a host intracellular compartment. Cell Host Microbe 2008, 3, 30–38. [Google Scholar] [CrossRef] [PubMed]
- Fowler, C.C.; Stack, G.; Jiao, X.Y.; Lara-Tejero, M.; Galan, J.E. Alternate subunit assembly diversifies the function of a bacterial toxin. Nat. Commun. 2019, 10, 624130. [Google Scholar] [CrossRef] [PubMed]
- Pons, B.J.; Vignard, J.; Mirey, G. Cytolethal distending toxin subunit B: A review of structure-function relationship. Toxins 2019, 11, 595. [Google Scholar] [CrossRef] [PubMed]
- DiRienzo, J.M. Uptake and processing of the cytolethal distending toxin by mammalian cells. Toxins 2014, 6, 3098–3116. [Google Scholar] [CrossRef]
- Frisan, T. Bacterial genotoxins: The long journey to the nucleus of mammalian cells. Biochim. Biophys. Acta 2016, 1858, 567–575. [Google Scholar] [CrossRef]
- Fowler, C.C.; Chang, S.J.; Gao, X.; Geiger, T.; Stack, G.; Galan, J.E. Emerging insights into the biology of typhoid toxin. Curr. Opin. Microbiol. 2017, 35, 70–77. [Google Scholar] [CrossRef]
- Fedor, Y.; Vignard, J.; Nicolau-Travers, M.L.; 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 the Escherichia coli Cytolethal Distending Toxin. Cell. Microbiol. 2013, 15, 1–15. [Google Scholar] [CrossRef]
- Sert, V.; Cans, C.; Tasca, C.; Bret-Bennis, L.; Oswald, E.; Ducommun, B.; De Rycke, J. The bacterial cytolethal distending toxin (CDT) triggers a G2 cell cycle checkpoint in mammalian cells without preliminary induction of DNA strand breaks. Oncogene 1999, 18, 6296–6304. [Google Scholar] [CrossRef]
- Comayras, C.; Tasca, C.; Peres, S.Y.; Ducommun, B.; Oswald, E.; De Rycke, J. Escherichia coli cytolethal distending toxin blocks the HeLa cell cycle at the G2/M transition by preventing cdc2 protein kinase dephosphorylation and activation. Infect. Immun. 1997, 65, 5088–5095. [Google Scholar] [CrossRef]
- Li, L.; Sharipo, A.; Chaves-Olarte, E.; Masucci, M.G.; Levitsky, V.; Thelestam, M.; Frisan, T. The Haemophilus ducreyi cytolethal distending toxin activates sensors of DNA damage and repair complexes in proliferating and non-proliferating cells. Cell. Micro. 2002, 4, 87–99. [Google Scholar] [CrossRef] [PubMed]
- Elwell, C.; Chao, K.; Patel, K.; Dreyfus, L. Escherichia coli CdtB mediates cytolethal distending toxin cell cycle arrest. Infect. Immun. 2001, 69, 3418–3422. [Google Scholar] [CrossRef] [PubMed]
- Dlakic, M. Is CdtB a nuclease or a phosphatase? Technical comment. Science 2001, 291, 547a. [Google Scholar] [CrossRef] [PubMed]
- Shenker, B.J.; Dlakic, M.; Walker, L.P.; Besack, D.; Jaffe, E.; LaBelle, E.; Boesze-Battaglia, K. A novel mode of action for a microbial-derived immunotoxin: The cytolethal distending toxin subunit B exhibits phosphatidylinositol 3,4,5-triphosphate phosphatase activity. J. Immunol. 2007, 178, 5099–5108. [Google Scholar] [CrossRef] [PubMed]
- Guerra, L.; Cortes-Bratti, X.; Guidi, R.; Frisan, T. The biology of the cytolethal distending toxins. Toxins 2011, 3, 172–190. [Google Scholar] [CrossRef] [PubMed]
- Guidi, R.; Guerra, L.; Levi, L.; Stenerlow, B.; Fox, J.G.; Josenhans, C.; Masucci, M.G.; Frisan, T. Chronic exposure to the cytolethal distending toxins of Gram-negative bacteria promotes genomic instability and altered DNA damage response. Cell. Microbiol. 2013, 15, 98–113. [Google Scholar] [CrossRef]
- Graillot, V.; Dormoy, I.; Dupuy, J.; Shay, J.W.; Huc, L.; Mirey, G.; Vignard, J. Genotoxicity of cytolethal distending toxin (CDT) on isogenic human colorectal cell lines: Potential promoting effects for colorectal carcinogenesis. Front. Cell. Infect. Microbiol. 2016, 6, 34. [Google Scholar] [CrossRef]
- Fox, J.G.; Rogers, A.B.; Whary, M.T.; Ge, Z.; Taylor, N.S.; Xu, S.; Horwitz, B.H.; Erdman, S.E. Gastroenteritis in NF-kappaB-deficient mice is produced with wild-type Camplyobacter jejuni but not with C. jejuni lacking cytolethal distending toxin despite persistent colonization with both strains. Infect. Immun. 2004, 72, 1116–1125. [Google Scholar] [CrossRef]
- Ge, Z.; Rogers, A.B.; Feng, Y.; Lee, A.; Xu, S.; Taylor, N.S.; Fox, J.G. Bacterial cytolethal distending toxin promotes the development of dysplasia in a model of microbially induced hepatocarcinogenesis. Cell. Microbiol. 2007, 9, 2070–2080. [Google Scholar] [CrossRef]
- He, Z.; Gharaibeh, R.Z.; Newsome, R.C.; Pope, J.; Dougherty, M.W.; Tomkovich, S.; Pons, B.; Mirey, G.; Vignard, J.; Hendrixson, D.R.; et al. Campylobacterjejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut 2018. [Google Scholar] [CrossRef]
- Buc, E.; Dubois, D.; Sauvanet, P.; Raisch, J.; Delmas, J.; Darfeuille-Michaud, A.; Pezet, D.; Bonnet, R. High prevalence of mucosa-associated E. coli producing cyclomodulin and genotoxin in colon cancer. PLoS ONE 2013, 8, e56964. [Google Scholar] [CrossRef] [PubMed]
- Dutta, U.; Garg, P.K.; Kumar, R.; Tandon, R.K. Typhoid carriers among patients with gallstones are at increased risk for carcinoma of the gallbladder. Am. J. Gastroenterol. 2000, 95, 784–787. [Google Scholar] [CrossRef] [PubMed]
- Nougayrede, J.P.; Homburg, S.; Taieb, F.; Boury, M.; Brzuszkiewicz, E.; Gottschalk, G.; Buchrieser, C.; Hacker, J.; Dobrindt, U.; Oswald, E. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science 2006, 313, 848–851. [Google Scholar] [CrossRef] [PubMed]
- Nowrouzian, F.L.; Oswald, E. Escherichia coli strains with the capacity for long-term persistence in the bowel microbiota carry the potentially genotoxic pks island. Microb. Pathog. 2012, 53, 180–182. [Google Scholar] [CrossRef]
- Arthur, J.C.; Perez-Chanona, E.; Muhlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123. [Google Scholar] [CrossRef]
- Prorok-Hamon, M.; Friswell, M.K.; Alswied, A.; Roberts, C.L.; Song, F.; Flanagan, P.K.; Knight, P.; Codling, C.; Marchesi, J.R.; Winstanley, C.; et al. Colonic mucosa-associated diffusely adherent afaC+ Escherichia coli expressing lpfA and pks are increased in inflammatory bowel disease and colon cancer. Gut 2014, 63, 761–770. [Google Scholar] [CrossRef]
- Eklof, V.; Lofgren-Burstrom, A.; Zingmark, C.; Edin, S.; Larsson, P.; Karling, P.; Alexeyev, O.; Rutegard, J.; Wikberg, M.L.; Palmqvist, R. Cancer-associated fecal microbial markers in colorectal cancer detection. Int. J. Cancer 2017, 141, 2528–2536. [Google Scholar] [CrossRef]
- Olier, M.; Marcq, I.; Salvador-Cartier, C.; Secher, T.; Dobrindt, U.; Boury, M.; Bacquie, V.; Penary, M.; Gaultier, E.; Nougayrede, J.P.; et al. Genotoxicity of Escherichia coli Nissle 1917 strain cannot be dissociated from its probiotic activity. Gut Microbes 2012, 3, 501–509. [Google Scholar] [CrossRef]
- Bossuet-Greif, N.; Vignard, J.; Taieb, F.; Mirey, G.; Dubois, D.; Petit, C.; Oswald, E.; Nougayrede, J.P. The colibactin genotoxin generates DNA interstrand cross-links in infected cells. mBio 2018, 9, e02397-17. [Google Scholar] [CrossRef]
- Fais, T.; Cougnoux, A.; Dalmasso, G.; Laurent, F.; Delmas, J.; Bonnet, R. Antibiotic activity of Escherichia coli against Multiresistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2016, 60, 6986–6988. [Google Scholar] [CrossRef][Green Version]
- Massip, C.; Branchu, P.; Bossuet-Grief, N.; Chagneau, C.V.; Gaillard, D.; Martin, P.; Boury, M.; Secher, T.; Dubois, D.; Nougayrede, J.P.; et al. Deciphering the interplay between the genotoxic and probiotic activities of Escherichia coli Nissle 1917. PLoS Pathog. 2019, 15, e1008029. [Google Scholar] [CrossRef] [PubMed]
- Perez-Berezo, T.; Pujo, J.; Martin, P.; Le Faouder, P.; Galano, J.M.; Guy, A.; Knauf, C.; Tabet, J.C.; Tronnet, S.; Barreau, F.; et al. Identification of an analgesic lipopeptide produced by the probiotic Escherichia coli strain Nissle 1917. Nat. Commun. 2017, 8, 1314. [Google Scholar] [CrossRef] [PubMed]
- Cuevas-Ramos, G.; Petit, C.R.; Marcq, I.; Boury, M.; Oswald, E.; Nougayrede, J.P. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc. Natl. Acad. Sci. USA 2010, 107, 11537–11542. [Google Scholar] [CrossRef] [PubMed]
- Payros, D.; Secher, T.; Boury, M.; Brehin, C.; Menard, S.; Salvador-Cartier, C.; Cuevas-Ramos, G.; Watrin, C.; Marcq, I.; Nougayrede, J.P.; et al. Maternally acquired genotoxic Escherichia coli alters offspring’s intestinal homeostasis. Gut Microbes 2014, 5, 313–325. [Google Scholar] [CrossRef] [PubMed]
- Secher, T.; Payros, D.; Brehin, C.; Boury, M.; Watrin, C.; Gillet, M.; Bernard-Cadenat, I.; Menard, S.; Theodorou, V.; Saoudi, A.; et al. Oral tolerance failure upon neonatal gut colonization with Escherichia coli producing the genotoxin colibactin. Infect. Immun. 2015, 83, 2420–2429. [Google Scholar] [CrossRef]
- Ciccia, A.; Elledge, S.J. The DNA damage response: Making it safe to play with knives. Mol. Cell 2010, 40, 179–204. [Google Scholar] [CrossRef]
- Blackford, A.N.; Jackson, S.P. ATM, ATR, and DNA-PK: The trinity at the heart of the DNA damage response. Mol. Cell 2017, 66, 801–817. [Google Scholar] [CrossRef]
- Nepal, M.; Che, R.; Zhang, J.; Ma, C.; Fei, P. Fanconi anemia signaling and cancer. Trends Cancer 2017, 3, 840–856. [Google Scholar] [CrossRef]
- Walden, H.; Deans, A.J. The Fanconi anemia DNA repair pathway: Structural and functional insights into a complex disorder. Annu. Rev. Biophys. 2014, 43, 257–278. [Google Scholar] [CrossRef]
- Che, R.; Zhang, J.; Nepal, M.; Han, B.; Fei, P. Multifaceted Fanconi anemia signaling. Trends Genet. 2018, 34, 171–183. [Google Scholar] [CrossRef]
- Mahaney, B.L.; Meek, K.; Lees-Miller, S.P. Repair of ionizing radiation-induced DNA double-strand breaks by non-homologous end-joining. Biochem. J. 2009, 417, 639–650. [Google Scholar] [CrossRef] [PubMed]
- Pannunzio, N.R.; Watanabe, G.; Lieber, M.R. Nonhomologous DNA end-joining for repair of DNA double-strand breaks. J. Biol. Chem. 2018, 293, 10512–10523. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Yu, X. The role of poly(ADP-ribosyl)ation in DNA damage response and cancer chemotherapy. Oncogene 2015, 34, 3349–3356. [Google Scholar] [CrossRef] [PubMed]
- Scully, R.; Panday, A.; Elango, R.; Willis, N.A. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat. Rev. Mol. Cell Biol. 2019, 20, 698–714. [Google Scholar] [CrossRef] [PubMed]
- Shiloh, Y.; Ziv, Y. The ATM protein kinase: Regulating the cellular response to genotoxic stress, and more. Nat. Rev. Mol. Cell Biol. 2013, 14, 197–210. [Google Scholar] [CrossRef]
- Rogakou, E.P.; Boon, C.; Redon, C.; Bonner, W.M. Megabase chromatin domains involved in DNA double-strand breaks in vivo. J. Cell Biol. 1999, 146, 905–916. [Google Scholar] [CrossRef]
- Zeman, M.K.; Cimprich, K.A. Causes and consequences of replication stress. Nat. Cell Biol. 2014, 16, 2–9. [Google Scholar] [CrossRef]
- Zou, L.; Elledge, S.J. Sensing DNA damage through ATRIP recognition of RPA-ssDNA complexes. Science 2003, 300, 1542–1548. [Google Scholar] [CrossRef]
- Bermudez, V.P.; Lindsey-Boltz, L.A.; Cesare, A.J.; Maniwa, Y.; Griffith, J.D.; Hurwitz, J.; Sancar, A. Loading of the human 9-1-1 checkpoint complex onto DNA by the checkpoint clamp loader hRad17-replication factor C complex in vitro. Proc. Natl. Acad. Sci. USA 2003, 100, 1633–1638. [Google Scholar] [CrossRef]
- Yazinski, S.A.; Zou, L. Functions, regulation, and therapeutic implications of the ATR checkpoint pathway. Annu. Rev. Genet. 2016, 50, 155–173. [Google Scholar] [CrossRef]
- Ma, M.; Rodriguez, A.; Sugimoto, K. Activation of ATR-related protein kinase upon DNA damage recognition. Curr. Genet. 2019. [Epub ahead of print]. [Google Scholar] [CrossRef] [PubMed]
- Stoepker, C.; Hain, K.; Schuster, B.; Hilhorst-Hofstee, Y.; Rooimans, M.A.; Steltenpool, J.; Oostra, A.B.; Eirich, K.; Korthof, E.T.; Nieuwint, A.W.; et al. SLX4, a coordinator of structure-specific endonucleases, is mutated in a new Fanconi anemia subtype. Nat. Genet. 2011, 43, 138–141. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Roos, W.P.; Thomas, A.D.; Kaina, B. DNA damage and the balance between survival and death in cancer biology. Nat. Rev. Cancer 2016, 16, 20–33. [Google Scholar] [CrossRef] [PubMed]
- Rodier, F.; Campisi, J. Four faces of cellular senescence. J. Cell Biol. 2011, 192, 547–556. [Google Scholar] [CrossRef] [PubMed]
- Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of cellular senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef]
- Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular senescence: Defining a path forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
- Krtolica, A.; Parrinello, S.; Lockett, S.; Desprez, P.Y.; Campisi, J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proc. Natl. Acad. Sci. USA 2001, 98, 12072–12077. [Google Scholar] [CrossRef]
- Campisi, J.; d’Adda di Fagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef]
- Ohanna, M.; Giuliano, S.; Bonet, C.; Imbert, V.; Hofman, V.; Zangari, J.; Bille, K.; Robert, C.; Bressac-de Paillerets, B.; Hofman, P.; et al. Senescent cells develop a PARP-1 and nuclear factor-{kappa}B-associated secretome (PNAS). Genes Dev. 2011, 25, 1245–1261. [Google Scholar] [CrossRef]
- Kastenhuber, E.R.; Lowe, S.W. Putting p53 in context. Cell 2017, 170, 1062–1078. [Google Scholar] [CrossRef] [PubMed]
- Kumari, R.; Kohli, S.; Das, S. p53 regulation upon genotoxic stress: Intricacies and complexities. Mol. Cell Oncol. 2014, 1, e969653. [Google Scholar] [CrossRef] [PubMed]
- Cortes-Bratti, X.; Karlsson, C.; Lagergard, T.; Thelestam, M.; Frisan, T. The Haemophilus ducreyi cytolethal distending toxin induces cell cycle arrest and apoptosis via the DNA damage checkpoint pathways. J. Biol. Chem. 2001, 276, 5296–5302. [Google Scholar] [CrossRef] [PubMed]
- Cortes-Bratti, X.; Chaves-Olarte, E.; Lagergård, T.; Thelestam, M. The cytolethal distending toxin from the chancroid bacterium Haemophilus ducreyi induces cell-cycle arrest in the G2 phase. J. Clin. Investig. 1999, 103, 107–115. [Google Scholar] [CrossRef]
- Escalas, N.; Davezac, N.; De Rycke, J.; Baldin, V.; Mazars, R.; Ducommun, B. Study of the cytolethal distending toxin-induced cell cycle arrest in HeLa cells: Involvement of the Cdc25 phosphatases. Exp. Cell Res. 2000, 257, 206–212. [Google Scholar] [CrossRef]
- Hassane, D.C.; Lee, R.B.; Pickett, C.L. Campylobacter jejuni cytolethal distending toxin promotes DNA repair responses in normal human cells. Infect. Immun. 2003, 71, 541–545. [Google Scholar] [CrossRef]
- Yamamoto, K.; Tominaga, K.; Sukedai, M.; Okinaga, T.; Iwanaga, K.; Nishihara, T.; Fukuda, J. Delivery of cytolethal distending toxin B induces cell cycle arrest and apoptosis in gingival squamous cell carcinoma in vitro. Eur. J. Oral Sci. 2004, 112, 445–451. [Google Scholar] [CrossRef]
- Sato, T.; Koseki, T.; Yamato, K.; Saiki, K.; Konishi, K.; Yoshikawa, M.; Ishikawa, I.; Nishihara, T. p53-independent expression of p21(CIP1/WAF1) in plasmacytic cells during G(2) cell cycle arrest induced by Actinobacillus actinomycetemcomitans cytolethal distending toxin. Infect. Immun. 2002, 70, 528–534. [Google Scholar] [CrossRef]
- Guerra, L.; Albihn, A.; Tronnersjö, S.; Yan, Q.; Guidi, R.; Stenerlöw, B.; Sterzenbach, T.; Josenhans, C.; Fox, J.G.; Schauer, D.B.; et al. Myc is required for activation of the ATM-dependent checkpoints in response to DNA damage. PLoS ONE 2010, 5, e892. [Google Scholar] [CrossRef]
- Blazkova, H.; Krejcikova, K.; Moudry, P.; Frisan, T.; Hodny, Z.; Bartek, J. Bacterial intoxication evokes cellular senescence with persistent DNA damage and cytokine signaling. J. Cell Mol. Med. 2010, 14, 357–367. [Google Scholar] [CrossRef]
- Fahrer, J.; Huelsenbeck, J.; Jaurich, H.; Dorsam, B.; Frisan, T.; Eich, M.; Roos, W.P.; Kaina, B.; Fritz, G. Cytolethal distending toxin (CDT) is a radiomimetic agent and induces persistent levels of DNA double-strand breaks in human fibroblasts. DNA Repair 2014, 18, 31–43. [Google Scholar] [CrossRef] [PubMed]
- Guidi, R.; Levi, L.; Rouf, S.F.; Puiac, S.; Rhen, M.; Frisan, T. Salmonella enterica delivers its genotoxin through outer membrane vesicles secreted from infected cells. Cell. Microbiol. 2013, 15, 2034–2050. [Google Scholar] [CrossRef] [PubMed]
- Bezine, E.; Malaise, Y.; Loeuillet, A.; Chevalier, M.; Boutet-Robinet, E.; Salles, B.; Mirey, G.; Vignard, J. Cell resistance to the cytolethal distending toxin involves an association of DNA repair mechanisms. Sci. Rep. UK 2016, 6, 36022. [Google Scholar] [CrossRef] [PubMed]
- Miller, R.A.; Wiedmann, M. The cytolethal distending toxin produced by nontyphoidal Salmonella serotypes Javiana, Montevideo, Oranienburg, and Mississippi induces DNA damage in a manner similar to that of serotype Typhi. mBio 2016, 7, e02109-16. [Google Scholar] [CrossRef] [PubMed]
- Ibler, A.E.M.; ElGhazaly, M.; Naylor, K.L.; Bulgakova, N.A.; El-Khamisy, S.F.; Humphreys, D. Typhoid toxin exhausts the RPA response to DNA replication stress driving senescence and Salmonella infection. Nat. Commun. 2019, 10, 4040. [Google Scholar] [CrossRef]
- Frisan, T.; Cortes-Bratti, X.; Chaves-Olarte, E.; Stenerlöw, B.; Thelestam, M. The Haemophilus ducreyi cytolethal distending toxin induces DNA double strand breaks and promotes ATM-dependent activation of RhoA. Cell. Micro. 2003, 5, 695–707. [Google Scholar] [CrossRef]
- Guerra, L.; Carr, H.S.; Richter-Dahlfors, A.; Masucci, M.G.; Thelestam, M.; Frost, J.A.; Frisan, T. A bacterial cytotoxin identifies the RhoA exchange factor Net1 as a key effector in the response to DNA damage. PLoS ONE 2008, 3, e2254. [Google Scholar] [CrossRef]
- Levi, L.; Toyooka, T.; Patarroyo, M.; Frisan, T. Bacterial genotoxins promote inside-out integrin beta1 activation, formation of focal adhesion complexes and cell spreading. PLoS ONE 2015, 10, e0124119. [Google Scholar] [CrossRef]
- Rankin, L.C.; Artis, D. Beyond host defense: Emerging functions of the immune system in regulating complex tissue physiology. Cell 2018, 173, 554–567. [Google Scholar] [CrossRef]
- Eberl, G. Immunity by equilibrium. Nat. Rev. Immunol. 2016, 16, 524–532. [Google Scholar] [CrossRef]
- Annunziato, F.; Romagnani, C.; Romagnani, S. The 3 major types of innate and adaptive cell-mediated effector immunity. J. Allergy Clin. Immunol. 2015, 135, 626–635. [Google Scholar] [CrossRef] [PubMed]
- Gasteiger, G.; Rudensky, A.Y. Interactions between innate and adaptive lymphocytes. Nat. Rev. Immunol. 2014, 14, 631–639. [Google Scholar] [CrossRef] [PubMed]
- Kotas, M.E.; Locksley, R.M. Why ILCs? Immunity 2018, 48, 1081–1090. [Google Scholar] [CrossRef] [PubMed]
- Klose, C.S.N.; Flach, M.; Mohle, L.; Rogell, L.; Hoyler, T.; Ebert, K.; Fabiunke, C.; Pfeifer, D.; Sexl, V.; Fonseca-Pereira, D.; et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 2014, 157, 340–356. [Google Scholar] [CrossRef] [PubMed]
- Roan, F.; Obata-Ninomiya, K.; Ziegler, S.F. Epithelial cell derived cytokines: More than just signaling the alarm. J. Clin. Investig. 2019, 129, 1441–1451. [Google Scholar] [CrossRef] [PubMed]
- Oyesola, O.O.; Fruh, S.P.; Webb, L.M.; Tait Wojno, E.D. Cytokines and beyond: Regulation of innate immune responses during helminth infection. Cytokine 2018, S1043-4666, 30356–30359. [Google Scholar] [CrossRef]
- Miossec, P.; Korn, T.; Kuchroo, V.K. Interleukin-17 and type 17 helper T cells. N. Engl. J. Med. 2009, 361, 888–898. [Google Scholar] [CrossRef]
- Matzinger, P.; Kamala, T. Tissue-based class control: The other side of tolerance. Nat. Rev. Immunol. 2011, 11, 221–230. [Google Scholar] [CrossRef]
- Janka, A.; Bielaszewska, M.; Dobrindt, U.; Greune, L.; Schmidt, M.A.; Karch, H. Cytolethal distending toxin gene cluster in enterohemorrhagic Escherichia coli O157:H- and O157:H7: Characterization and evolutionary considerations. Infect. Immun. 2003, 71, 3634–3638. [Google Scholar] [CrossRef]
- Peres, S.Y.; Marches, O.; Daigle, F.; Nougayrede, J.P.; Herault, F.; Tasca, C.; De Rycke, J.; Oswald, E. A new cytolethal distending toxin (CDT) from Escherichia coli producing CNF2 blocks HeLa cell division in G2/M phase. Mol. Microbiol. 1997, 24, 1095–1107. [Google Scholar] [CrossRef]
- Allue-Guardia, A.; Imamovic, L.; Muniesa, M. Evolution of a self-inducible cytolethal distending toxin type V-encoding bacteriophage from Escherichia coli O157:H7 to Shigella sonnei. J. Virol. 2013, 87, 13665–13675. [Google Scholar] [CrossRef] [PubMed][Green Version]
- Tilg, H.; Adolph, T.E.; Gerner, R.R.; Moschen, A.R. The intestinal microbiota in colorectal cancer. Cancer Cell 2018, 33, 954–964. [Google Scholar] [CrossRef] [PubMed]
- Van Elsland, D.; Neefjes, J. Bacterial infections and cancer. EMBO Rep. 2018, 19, e46632. [Google Scholar] [CrossRef] [PubMed]
- Akifusa, S.; Poole, S.; Lewthwaite, J.; Henderson, B.; Nair, S.P. Recombinant Actinobacillus actinomycetemcomitans cytolethal distending toxin proteins are required to interact to inhibit human cell cycle progression and to stimulate human leukocyte cytokine synthesis. Infect. Immun. 2001, 69, 5925–5930. [Google Scholar] [CrossRef]
- Shenker, B.J.; Walker, L.P.; Zekavat, A.; Dlakic, M.; Boesze-Battaglia, K. Blockade of the PI-3K signalling pathway by the Aggregatibacter actinomycetemcomitans cytolethal distending toxin induces macrophages to synthesize and secrete pro-inflammatory cytokines. Cell. Microbiol. 2014, 16, 1391–1404. [Google Scholar] [CrossRef]
- Shenker, B.J.; Ojcius, D.M.; Walker, L.P.; Zekavat, A.; Scuron, M.D.; Boesze-Battaglia, K. Aggregatibacter actinomycetemcomitans cytolethal distending toxin activates the NLRP3 inflammasome in human macrophages, leading to the release of proinflammatory cytokines. Infect. Immun. 2015, 83, 1487–1496. [Google Scholar] [CrossRef]
- Ando-Suguimoto, E.S.; da Silva, M.P.; Kawamoto, D.; Chen, C.; DiRienzo, J.M.; Mayer, M.P. The cytolethal distending toxin of Aggregatibacter actinomycetemcomitans inhibits macrophage phagocytosis and subverts cytokine production. Cytokine 2014, 66, 46–53. [Google Scholar] [CrossRef]
- Belibasakis, G.N.; Johansson, A.; Wang, Y.; Chen, C.; Lagergard, T.; Kalfas, S.; Lerner, U.H. Cytokine responses of human gingival fibroblasts to Actinobacillus actinomycetemcomitans cytolethal distending toxin. Cytokine 2005, 30, 56–63. [Google Scholar] [CrossRef]
- Hickey, T.E.; McVeigh, A.L.; Scott, D.A.; Michielutti, R.E.; Bixby, A.; Carroll, S.A.; Bourgeois, A.L.; Guerry, P. Campylobacter jejuni cytolethal distending toxin mediates release of interleukin-8 from intestinal epithelial cells. Infect. Immun. 2000, 68, 6535–6541. [Google Scholar] [CrossRef]
- Zheng, J.; Meng, J.; Zhao, S.; Singh, R.; Song, W. Campylobacter-induced interleukin-8 secretion in polarized human intestinal epithelial cells requires Campylobacter-secreted cytolethal distending toxin- and Toll-like receptor-mediated activation of NF-kappaB. Infect. Immun. 2008, 76, 4498–4508. [Google Scholar] [CrossRef]
- Pere-Vedrenne, C.; Cardinaud, B.; Varon, C.; Mocan, I.; Buissonniere, A.; Izotte, J.; Megraud, F.; Menard, A. The cytolethal distending toxin subunit CdtB of helicobacter induces a Th17-related and antimicrobial signature in intestinal and hepatic cells in vitro. J. Infect. Dis. 2016, 213, 1979–1989. [Google Scholar] [CrossRef] [PubMed]
- Buchmann, K. Evolution of innate immunity: Clues from invertebrates via fish to mammals. Front. Immunol. 2014, 5, 459. [Google Scholar] [CrossRef] [PubMed]
- McCool, K.W.; Miyamoto, S. DNA damage-dependent NF-kappaB activation: NEMO turns nuclear signaling inside out. Immunol. Rev. 2012, 246, 311–326. [Google Scholar] [CrossRef] [PubMed]
- Tak, P.P.; Firestein, G.S. NF-kappa B: A key role in inflammatory diseases. J. Clin. Investig. 2001, 107, 7–11. [Google Scholar] [CrossRef]
- Cuenda, A.; Rousseau, S. p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim. Biophys. Acta 2007, 1773, 1358–1375. [Google Scholar] [CrossRef]
- Di Maggio, F.M.; Minafra, L.; Forte, G.I.; Cammarata, F.P.; Lio, D.; Messa, C.; Gilardi, M.C.; Bravata, V. Portrait of inflammatory response to ionizing radiation treatment. J. Inflamm. 2015, 12, 14. [Google Scholar] [CrossRef]
- Harding, S.M.; Benci, J.L.; Irianto, J.; Discher, D.E.; Minn, A.J.; Greenberg, R.A. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 2017, 548, 466–470. [Google Scholar] [CrossRef]
- Rodier, F.; Coppe, J.P.; Patil, C.K.; Hoeijmakers, W.A.; Munoz, D.P.; Raza, S.R.; Freund, A.; Campeau, E.; Davalos, A.R.; Campisi, J. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 2009, 11, 973–979. [Google Scholar] [CrossRef]
- Ahn, J.; Gutman, D.; Saijo, S.; Barber, G.N. STING manifests self DNA-dependent inflammatory disease. Proc. Natl. Acad. Sci. USA 2012, 109, 19386–19391. [Google Scholar] [CrossRef]
- Lan, Y.Y.; Londono, D.; Bouley, R.; Rooney, M.S.; Hacohen, N. Dnase2a deficiency uncovers lysosomal clearance of damaged nuclear DNA via autophagy. Cell Rep. 2014, 9, 180–192. [Google Scholar] [CrossRef]
- Hartlova, A.; Erttmann, S.F.; Raffi, F.A.; Schmalz, A.M.; Resch, U.; Anugula, S.; Lienenklaus, S.; Nilsson, L.M.; Kroger, A.; Nilsson, J.A.; et al. DNA damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 2015, 42, 332–343. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Chen, Z.J. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 2018, 215, 1287–1299. [Google Scholar] [CrossRef] [PubMed]
- Kondo, T.; Kobayashi, J.; Saitoh, T.; Maruyama, K.; Ishii, K.J.; Barber, G.N.; Komatsu, K.; Akira, S.; Kawai, T. DNA damage sensor MRE11 recognizes cytosolic double-stranded DNA and induces type I interferon by regulating STING trafficking. Proc. Natl. Acad. Sci. USA 2013, 110, 2969–2974. [Google Scholar] [CrossRef] [PubMed]
- Martin, O.C.B.; Bergonzini, A.; D’Amico, F.; Chen, P.; Shay, J.W.; Dupuy, J.; Svensson, M.; Masucci, M.G.; Frisan, T. Infection with genotoxin-producing Salmonella enterica synergises with loss of the tumour suppressor APC in promoting genomic instability via the PI3K pathway in colonic epithelial cells. Cell. Microbiol. 2019, 21, e13099. [Google Scholar] [CrossRef]
- Secher, T.; Samba-Louaka, A.; Oswald, E.; Nougayrede, J.P. Escherichia coli producing colibactin triggers premature and transmissible senescence in mammalian cells. PLoS ONE 2013, 8, e77157. [Google Scholar] [CrossRef]
- Cougnoux, A.; Dalmasso, G.; Martinez, R.; Buc, E.; Delmas, J.; Gibold, L.; Sauvanet, P.; Darcha, C.; Dechelotte, P.; Bonnet, M.; et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 2014, 63, 1932–1942. [Google Scholar] [CrossRef]
- Pratt, J.S.; Sachen, K.L.; Wood, H.D.; Eaton, K.A.; Young, V.B. Modulation of host immune responses by the cytolethal distending toxin of Helicobacter hepaticus. Infect. Immun. 2006, 74, 4496–4504. [Google Scholar] [CrossRef]
- Shen, Z.; Feng, Y.; Rogers, A.B.; Rickman, B.; Whary, M.T.; Xu, S.; Clapp, K.M.; Boutin, S.R.; Fox, J.G. Cytolethal distending toxin promotes Helicobacter cinaedi-associated typhlocolitis in interleukin-10-deficient mice. Infect. Immun. 2009, 77, 2508–2516. [Google Scholar] [CrossRef]
- Young, V.B.; Knox, K.A.; Pratt, J.S.; Cortez, J.S.; Mansfield, L.S.; Rogers, A.B.; Fox, J.G.; Schauer, D.B. In vitro and in vivo characterization of Helicobacter hepaticus cytolethal distending toxin mutants. Infect. Immun. 2004, 72, 2521–2527. [Google Scholar] [CrossRef]
- Ge, Z.; Feng, Y.; Whary, M.T.; Nambiar, P.R.; Xu, S.; Ng, V.; Taylor, N.S.; Fox, J.G. Cytolethal distending toxin is essential for Helicobacter hepaticus colonization in outbred Swiss Webster mice. Infect. Immun. 2005, 73, 3559–3567. [Google Scholar] [CrossRef]
- Ge, Z.; Feng, Y.; Ge, L.; Parry, N.; Muthupalani, S.; Fox, J.G. Helicobacter hepaticus cytolethal distending toxin promotes intestinal carcinogenesis in 129Rag2-deficient mice. Cell. Microbiol. 2017, 19, e12728. [Google Scholar] [CrossRef]
- Shenker, B.J.; McKay, T.; Datar, S.; Miller, M.; Chowhan, R.; Demuth, D. Actinobacillus actinomycetemcomitans immunosuppressive protein is a member of the family of cytolethal distending toxins capable of causing a G2 arrest in human T cells. J. Immunol. 1999, 162, 4773–4780. [Google Scholar]
- Shenker, B.J.; Hoffmaster, R.H.; Zekavat, A.; Yamaguchi, N.; Lally, E.T.; Demuth, D.R. Induction of apoptosis in human T cells by Actinobacillus actinomycetemcomitans cytolethal distending toxin is a consequence of G2 arrest of the cell cycle. J. Immunol. 2001, 167, 435–441. [Google Scholar] [CrossRef] [PubMed]
- Ohara, M.; Hayashi, T.; Kusunoki, Y.; Miyauchi, M.; Takata, T.; Sugai, M. Caspase-2 and caspase-7 are involved in cytolethal distending toxin-induced apoptosis in Jurkat and MOLT-4 T-cell lines. Infect. Immun. 2004, 72, 871–879. [Google Scholar] [CrossRef] [PubMed]
- Ohara, M.; Hayashi, T.; Kusunoki, Y.; Nakachi, K.; Fujiwara, T.; Komatsuzawa, H.; Sugai, M. Cytolethal distending toxin induces caspase-dependent and -independent cell death in MOLT-4 cells. Infect. Immun. 2008, 76, 4783–4791. [Google Scholar] [CrossRef] [PubMed]
- Marcq, I.; Martin, P.; Payros, D.; Cuevas-Ramos, G.; Boury, M.; Watrin, C.; Nougayrede, J.P.; Olier, M.; Oswald, E. The genotoxin colibactin exacerbates lymphopenia and decreases survival rate in mice infected with septicemic Escherichia coli. J. Infect. Dis. 2014, 210, 285–294. [Google Scholar] [CrossRef] [PubMed]
- Gelfanova, V.; Hansen, E.J.; Spinola, S.M. Cytolethal distending toxin of Haemophilus ducreyi induces apoptotic death of Jurkat T cells. Infect. Immun. 1999, 67, 6394–6402. [Google Scholar] [PubMed]
- Svensson, L.; Tarkowski, A.; Thelestam, M.; Lagergård, T. The impact of Haemophilus ducreyi cytolethal distending toxin on cells involved in immune response. Microb. Pathog. 2001, 30, 157–166. [Google Scholar] [CrossRef] [PubMed]
- Wising, C.; Azem, J.; Zetterberg, M.; Svensson, L.A.; Ahlman, K.; Lagergard, T. Induction of apoptosis/necrosis in various human cell lineages by Haemophilus ducreyi cytolethal distending toxin. Toxicon Off. J. Int. Soc. Toxinol. 2005, 45, 767–776. [Google Scholar] [CrossRef]
- Del Bel Belluz, L.; Guidi, R.; Pateras, I.S.; Levi, L.; Mihaljevic, B.; Rouf, S.F.; Wrande, M.; Candela, M.; Turroni, S.; Nastasi, C.; et al. The typhoid toxin promotes host survival and the establishment of a persistent asymptomatic infection. PLoS Pathog. 2016, 12, e1005528. [Google Scholar] [CrossRef]
- Miller, R.A.; Betteken, M.I.; Guo, X.; Altier, C.; Duhamel, G.E.; Wiedmann, M. The typhoid toxin produced by the nontyphoidal Salmonella enterica serotype javiana is required for induction of a DNA damage response in vitro and systemic spread in vivo. mBio 2018, 9, e00467-18. [Google Scholar] [CrossRef] [PubMed]
- Westbrook, A.M.; Schiestl, R.H. Atm-deficient mice exhibit increased sensitivity to dextran sulfate sodium-induced colitis characterized by elevated DNA damage and persistent immune activation. Cancer Res. 2010, 70, 1875–1884. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Shen, Y.; Oishi, H.; Matteson, E.L.; Tian, L.; Goronzy, J.J.; Weyand, C.M. Restoring oxidant signaling suppresses proarthritogenic T cell effector functions in rheumatoid arthritis. Sci. Transl. Med. 2016, 8, 331ra38. [Google Scholar] [CrossRef] [PubMed]
- Figueiredo, N.; Chora, A.; Raquel, H.; Pejanovic, N.; Pereira, P.; Hartleben, B.; Neves-Costa, A.; Moita, C.; Pedroso, D.; Pinto, A.; et al. Anthracyclines induce DNA damage response-mediated protection against severe sepsis. Immunity 2013, 39, 874–884. [Google Scholar] [CrossRef]
- Smythies, L.E.; Sellers, M.; Clements, R.H.; Mosteller-Barnum, M.; Meng, G.; Benjamin, W.H.; Orenstein, J.M.; Smith, P.D. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J. Clin. Investig. 2005, 115, 66–75. [Google Scholar] [CrossRef]
- Denning, T.L.; Wang, Y.C.; Patel, S.R.; Williams, I.R.; Pulendran, B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat. Immunol. 2007, 8, 1086–1094. [Google Scholar] [CrossRef]
- Tanoue, T.; Atarashi, K.; Honda, K. Development and maintenance of intestinal regulatory T cells. Nat. Rev. Immunol. 2016, 16, 295–309. [Google Scholar] [CrossRef]
- Toso, A.; Revandkar, A.; Di Mitri, D.; Guccini, I.; Proietti, M.; Sarti, M.; Pinton, S.; Zhang, J.; Kalathur, M.; Civenni, G.; et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 2014, 9, 75–89. [Google Scholar] [CrossRef]
- Payros, D.; Dobrindt, U.; Martin, P.; Secher, T.; Bracarense, A.P.; Boury, M.; Laffitte, J.; Pinton, P.; Oswald, E.; Oswald, I.P. The food contaminant deoxynivalenol exacerbates the genotoxicity of gut microbiota. mBio 2017, 8, e00007-17. [Google Scholar] [CrossRef]
- Johnson, W.M.; Lior, H. Production of Shiga toxin and a cytolethal distending toxin (CLDT) by serogroups of Shigella spp. FEMS Microbiol. Lett. 1987, 48, 235–238. [Google Scholar] [CrossRef]
- Johnson, W.M.; Lior, H. Response of Chinese hamster ovary cells to a cytolethal distending toxin (CDT) of Escherichia coli and possible misinterpretation as heat-labile (LT) enterotoxin. FEMS Microbiol. Lett. 1987, 43, 19–23. [Google Scholar] [CrossRef]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Martin, O.C.B.; Frisan, T. Bacterial Genotoxin-Induced DNA Damage and Modulation of the Host Immune Microenvironment. Toxins 2020, 12, 63. https://doi.org/10.3390/toxins12020063
Martin OCB, Frisan T. Bacterial Genotoxin-Induced DNA Damage and Modulation of the Host Immune Microenvironment. Toxins. 2020; 12(2):63. https://doi.org/10.3390/toxins12020063
Chicago/Turabian StyleMartin, Océane C.B., and Teresa Frisan. 2020. "Bacterial Genotoxin-Induced DNA Damage and Modulation of the Host Immune Microenvironment" Toxins 12, no. 2: 63. https://doi.org/10.3390/toxins12020063
APA StyleMartin, O. C. B., & Frisan, T. (2020). Bacterial Genotoxin-Induced DNA Damage and Modulation of the Host Immune Microenvironment. Toxins, 12(2), 63. https://doi.org/10.3390/toxins12020063