Next Article in Journal
In Vitro and In Silico Analysis of the Inhibitory Activity of EGCG-Stearate against Herpes Simplex Virus-2
Previous Article in Journal
Metagenomic Analysis of the Species Composition and Seasonal Distribution of Marine Dinoflagellate Communities in Four Korean Coastal Regions
Previous Article in Special Issue
Novel Template Plasmids pCyaA’-Kan and pCyaA’-Cam for Generation of Unmarked Chromosomal cyaA’ Translational Fusion to T3SS Effectors in Salmonella
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue: Type III Secretion Systems in Human/Animal Pathogenic Bacteria

by
Joaquín Bernal-Bayard
and
Francisco Ramos-Morales
*
Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
*
Author to whom correspondence should be addressed.
Microorganisms 2022, 10(7), 1461; https://doi.org/10.3390/microorganisms10071461
Submission received: 6 July 2022 / Accepted: 12 July 2022 / Published: 20 July 2022
(This article belongs to the Special Issue Type III Secretion Systems in Human/Animal Pathogenic Bacteria)
Type III secretion systems (T3SSs) are molecular devices that are essential for the communication of many Gram-negative bacteria with their eukaryotic hosts [1]. They were initially described, some 30 years ago, in bacteria from the genera Yersinia and Salmonella [2,3], but are also present in many other Gram-negative bacteria including Aeromonas, Bordetella, Burkholderia, Chlamydia, Citrobacter, Edwardsiella, Erwinia, Escherichia, Photorhabdus, Pseudomonas, Ralstonia, Rhizobium, Shigella, Vibrio and Xanthomonas [4,5].
The structure of these multiprotein molecular machines, also known as injectisomes, has been studied in detail. It contains more than 20 proteins and includes a cytoplasmic sorting platform, an inner membrane export apparatus, a basal body, a needle complex, and a translocation pore in the host cell membrane [6,7,8,9,10].
At least eight subfamilies of injectisomes can be distinguished: Ysc, SPI-1, SPI-2, and the Chlamydia subfamily, which are present predominantly in animal pathogens; Hrp1, Hrp2, and the Rhizobium subfamily, present in plant pathogens; and the Myxococcus subfamily [11].
Using these systems bacterial pathogens and symbionts are able to inject proteins, known as effectors, into the cytosol of animal, plant, fungi, or protist cells [12]. These effectors may interact with specific components of the host to alter their behavior and interfere with signal transduction pathways to establish an appropriate survival niche. Although the physical interaction with a host target may be the main mechanism of action for some effectors, many effectors possess enzymatic activities that include guanine nucleotide activating, cysteine protease, acyltransferase, deubiquitinase, kinase, AND-ribosyl transferase, phosphothreonine lyase, zinc metalloprotease, glycosyltransferase, and ubiquitin ligase [13].
This Special Issue focuses on the study of type III secretion systems in animal pathogenic bacteria. Despite the variety of lifestyles and diseases caused by these pathogens, there are also common themes arising from the fact that they share the use of T3SSs as key virulence mechanisms, and that effectors from different bacteria may have similar designs, biochemical activities, and/or functions. Therefore, researchers working in a particular T3SS can benefit from the studies carried out on other bacteria.
The Special Issue “Type III secretion systems in human/animal pathogenic bacteria” gathers nine papers covering studies in bacteria from the genera Aeromonas, Citrobacter, Edwardsiella, Escherichia, Salmonella, and Shigella.
Working on Edwardsiellla ictaluri, which causes enteric septicemia of catfish, Dubytska and Thune [14] described in detail the development of the Edwardsiella containing vacuole (ECV) in infected macrophages. Immunofluorescence of vacuolar membrane markers shows that, although the recruitment of the early endosomal markers Rab5 and EEA1 and the late endosomal marker Rab7 is similar in ECVs from a wild-type strain and a T3SS-mutant strain, the differential colocalization of Lamp1 indicates that the T3SS is essential to prevent phagosomal/lysosomal fusion. This secretion system is also important to suppress autophagy and apoptosis in macrophages.
Barkalita et al. [15] developed a reporter system based on PhoA activity to monitor secretion through the injectisome of enteropathogenic Escherichia coli. Pendergrass et al. [16] describe another reporter system based on the secretion of the enzyme carboxypeptidase G2 that, after delivery cleaves the substrate Glu-CyFur, releases the fluorescent molecule CyFur, and results in a visible color change from yellow to purple. These authors show that this system is useful to detect T3SS inhibitors.
The fish pathogen Aeromonas salmonicida possesses a T3SS that is essential for its virulence and is encoded in the plasmid pAsa5. The region encoding this system may be lost during growth at 25 °C due to recombination between insertion sequences present on the plasmid, but there is great variability between strains for this phenomenon. A paper in this Special Issue by Marcoux et al. explored this variability in 50 strains of A. salmonicida subsp. salmonicida and performed a comparative genomic analysis to identify genes related to the molecular mechanism involved [17].
Single cell analysis is also represented in this Issue with a study carried out by Sánchez-Romero and Casadesús in Salmonella enterica that allowed the identification of four subpopulations depending on the expression of Salmonella pathogenicity island I and/or the flagellar regulon [18].
Fernández et al. [19] report the construction of plasmids that are useful to generate chromosomal CyaA’ translational fusions by homologous recombination using the Red recombination system. These fusions can be used to measure the level of expression of a gene and the secretion of a fusion to the culture supernatant by Western blot using anti-CyaA’ antibodies. Also, since CyaA’ from Bordetella pertussis is a calmodulin-dependent adenylate cyclase that is only active in eukaryotic cells, these fusions can be used to detect the translocation of an effector into host cells.
These research articles are complemented by three review papers. A review by Araujo-Garrido et al. [20] focuses on a family of effectors with arginine N-glycosyltransferase activity. This family includes NleB from Citrobacter rodentium, NleB1 and NleB2 from enteropathogenic and enterohemorrhagic E. coli, and SseK1, SseK2, and SseK3 from S. enterica.
Finally, the T3SS of Shigella is reviewed by two leading groups in the field. In addition, to describe the structure, assembly, and function of the system, both groups emphasize different aspects related to it. Muthuramalingam et al. [21] highlight the possibility of using the T3SS as a therapeutic target for small molecules or vaccines. Bajunaid et al. [22] focus on the role of the system in the escape of Shigella from the vacuole.
Altogether, the papers of this Special Issue give to interested readers an idea of the diversity and common themes of these fascinating nanomachines in animal pathogens.

Funding

Research in the laboratory of the authors is funded by the Spanish Ministerio de Ciencia e Innovación—Agencia Estatal de Investigación PID2019-106132RB-I00/AEI/10.13039/501100011033; Consejería de Transformación Económica, Industria, Conocimiento y Universidades, Junta de Andalucía and ERDF A way of making Europe, grant number P20_00576; and by Universidad de Sevilla, Consejería de Transformación Económica, Industria, Conocimiento y Universidades, Junta de Andalucía, and ERDF A way of making Europe, grant number US-1380805.

Acknowledgments

We thank the authors, the reviewers, and the staff of the editorial office of Microorganisms for their contributions and support to this Special Issue.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lara-Tejero, M.; Galán, J.E. The Injectisome, a Complex Nanomachine for Protein Injection into Mammalian Cells. EcoSal Plus 2019, 8, 245–259. [Google Scholar] [CrossRef] [PubMed]
  2. Michiels, T.; Wattiau, P.; Brasseur, R.; Ruysschaert, J.M.; Cornelis, G. Secretion of Yop proteins by Yersiniae. Infect. Immun. 1990, 58, 2840–2849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Galán, J.E.; Curtiss, R. Cloning and molecular characterization of genes whose products allow Salmonella typhimurium to penetrate tissue culture cells. Proc. Natl. Acad. Sci. USA 1989, 86, 6383–6387. [Google Scholar] [CrossRef] [Green Version]
  4. Bastedo, D.P.; Lo, T.; Laflamme, B.; Desveaux, D.; Guttman, D.S. Diversity and evolution of type III secreted effectors: A case study of three families. In Current Topics in Microbiology and Immunology; Springer Science and Business Media Deutschland GmbH: Berlin, Germany, 2020; Volume 427, pp. 201–230. [Google Scholar]
  5. Galán, J.E.; Collmer, A. Type III secretion machines: Bacterial devices for protein delivery into host cells. Science 1999, 284, 1322–1328. [Google Scholar] [CrossRef] [Green Version]
  6. Picking, W.D.; Barta, M.L. The Tip Complex: From Host Cell Sensing to Translocon Formation. Curr. Top. Microbiol. Immunol. 2020, 427, 173–199. [Google Scholar] [CrossRef] [PubMed]
  7. Dey, S.; Chakravarty, A.; Guha Biswas, P.; De Guzman, R.N. The type III secretion system needle, tip, and translocon. Protein Sci. 2019, 28, 1582–1593. [Google Scholar] [CrossRef]
  8. Miletic, S.; Goessweiner-Mohr, N.; Marlovits, T.C. The Structure of the Type III Secretion System Needle Complex. Curr. Top. Microbiol. Immunol. 2020, 427, 67–90. [Google Scholar] [CrossRef]
  9. Habenstein, B.; El Mammeri, N.; Tolchard, J.; Lamon, G.; Tawani, A.; Berbon, M.; Loquet, A. Structures of Type III Secretion System Needle Filaments. Curr. Top. Microbiol. Immunol. 2020, 427, 109–131. [Google Scholar] [CrossRef]
  10. Umrekar, T.R.; Cohen, E.; Drobnič, T.; Gonzalez-Rodriguez, N.; Beeby, M. CryoEM of bacterial secretion systems: A primer for microbiologists. Mol. Microbiol. 2021, 115, 366–382. [Google Scholar] [CrossRef]
  11. Diepold, A.; Armitage, J.P. Type III secretion systems: The bacterial flagellum and the injectisome. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2015, 370, 20150020. [Google Scholar] [CrossRef] [Green Version]
  12. Puhar, A.; Sansonetti, P.J. Type III secretion system. Curr. Biol. 2014, 24, R784–R791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Jennings, E.; Thurston, T.L.M.; Holden, D.W. Salmonella SPI-2 Type III Secretion System Effectors: Molecular Mechanisms and Physiological Consequences. Cell Host Microbe 2017, 22, 217–231. [Google Scholar] [CrossRef] [PubMed]
  14. Dubytska, L.P.; Thune, R.L. Early Intracellular Trafficking and Subsequent Activity of Programmed Cell Death in Channel Catfish Macrophages Infected with Edwardsiella ictaluri. Microorganisms 2020, 8, 1649. [Google Scholar] [CrossRef] [PubMed]
  15. Barkalita, L.; Portaliou, A.G.; Loos, M.S.; Yuan, B.; Karamanou, S.; Economou, A. A Reporter System for Fast Quantitative Monitoring of Type 3 Protein Secretion in Enteropathogenic E. coli. Microorganisms 2020, 8, 1786. [Google Scholar] [CrossRef]
  16. Pendergrass, H.A.; Johnson, A.L.; Hotinger, J.A.; May, A.E. Fluorescence Detection of Type III Secretion Using a Glu-CyFur Reporter System in Citrobacter rodentium. Microorganisms 2020, 8, 1953. [Google Scholar] [CrossRef]
  17. Marcoux, P.-É.; Vincent, A.T.; Massicotte, M.-A.; Paquet, V.E.; Doucet, É.J.; Hosseini, N.; Trudel, M.V.; Byatt, G.; Laurent, M.; Frenette, M.; et al. Systematic Analysis of the Stress-Induced Genomic Instability of Type Three Secretion System in Aeromonas salmonicida subsp. Salmonicida. Microorganisms 2020, 9, 85. [Google Scholar] [CrossRef]
  18. Sánchez-Romero, M.A.; Casadesús, J. Single Cell Analysis of Bistable Expression of Pathogenicity Island 1 and the Flagellar Regulon in Salmonella enterica. Microorganisms 2021, 9, 210. [Google Scholar] [CrossRef]
  19. Fernández, P.A.; Zabner, M.; Ortega, J.; Morgado, C.; Amaya, F.; Vera, G.; Rubilar, C.; Salas, B.; Cuevas, V.; Valenzuela, C.; et al. Novel Template Plasmids pCyaA’-Kan and pCyaA’-Cam for Generation of Unmarked Chromosomal cyaA’ Translational Fusion to T3SS Effectors in Salmonella. Microorganisms 2021, 9, 475. [Google Scholar] [CrossRef]
  20. Araujo-Garrido, J.L.; Bernal-Bayard, J.; Ramos-Morales, F. Type III Secretion Effectors with Arginine N-Glycosyltransferase Activity. Microorganisms 2020, 8, 357. [Google Scholar] [CrossRef]
  21. Muthuramalingam, M.; Whittier, S.K.; Picking, W.L.; Picking, W.D. The Shigella Type III Secretion System: An Overview from Top to Bottom. Microorganisms 2021, 9, 451. [Google Scholar] [CrossRef]
  22. Bajunaid, W.; Haidar-Ahmad, N.; Kottarampatel, A.H.; Manigat, F.O.; Silué, N.; Tchagang, C.F.; Tomaro, K.; Campbell-Valois, F.X. The T3SS of Shigella: Expression, Structure, Function, and Role in Vacuole Escape. Microorganisms 2020, 8, 1933. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bernal-Bayard, J.; Ramos-Morales, F. Special Issue: Type III Secretion Systems in Human/Animal Pathogenic Bacteria. Microorganisms 2022, 10, 1461. https://doi.org/10.3390/microorganisms10071461

AMA Style

Bernal-Bayard J, Ramos-Morales F. Special Issue: Type III Secretion Systems in Human/Animal Pathogenic Bacteria. Microorganisms. 2022; 10(7):1461. https://doi.org/10.3390/microorganisms10071461

Chicago/Turabian Style

Bernal-Bayard, Joaquín, and Francisco Ramos-Morales. 2022. "Special Issue: Type III Secretion Systems in Human/Animal Pathogenic Bacteria" Microorganisms 10, no. 7: 1461. https://doi.org/10.3390/microorganisms10071461

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop