gms | German Medical Science

Infektiologie Update 2018: 26. Jahrestagung der Paul-Ehrlich-Gesellschaft für Chemotherapie (PEG)

Paul-Ehrlich-Gesellschaft für Chemotherapie (PEG)

04. - 06.10.2018, Wien, Österreich

“AmiA is a Penicillin Target Enzyme with Dual Activity in the Intracellular Pathogen Chlamydia pneumoniae”

Meeting Abstract

  • A. Klöckner - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany; MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
  • C. Otten - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany
  • A. Derouaux - The Centre for Bacterial Cell Biology, Institute for Cell and Molecular Biosciences, Newcastle University, United Kingdom; Centre d'Ingénierie des Protéines, Université de Liège, Belgium
  • W. Vollmer - The Centre for Bacterial Cell Biology, Institute for Cell and Molecular Biosciences, Newcastle University, United Kingdom
  • H. Bühl - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany
  • S. De Benedetti - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany
  • D. Münch - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany; AirCuris GmbH & Co. KG, Wuppertal, Germany
  • M. Josten - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany
  • K. Mölleken - Institute of Functional Microbial Genomics, University of Düsseldorf, Germany
  • H. G. Sahl - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany
  • B. Henrichfreise - Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), Pharmaceutical Microbiology, University of Bonn, Germany

Infektiologie Update 2018. 26. Jahrestagung der Paul-Ehrlich-Gesellschaft für Chemotherapie (PEG). Wien, 04.-06.10.2018. Düsseldorf: German Medical Science GMS Publishing House; 2018. Doc18peg17

doi: 10.3205/18peg17, urn:nbn:de:0183-18peg171

Veröffentlicht: 8. Oktober 2018

© 2018 Klöckner et al.
Dieser Artikel ist ein Open-Access-Artikel und steht unter den Lizenzbedingungen der Creative Commons Attribution 4.0 License (Namensnennung). Lizenz-Angaben siehe http://creativecommons.org/licenses/by/4.0/.


Gliederung

Text

The discovery of penicillin initiated the golden age of antibiotic discovery. Antibiotics are central to the treatment and prevention of bacterial infections and have underpinned advances in cancer chemotherapy, surgery, the survival of pre-term infants and organ transplantation. Penicillin belongs to the beta-lactam antibiotics, one of the most prescribed antibiotic class. Nevertheless, the effects, besides inhibiting the penicillin-binding proteins, remains to be elucidated. In this work, we used chlamydiae as a model organism to get insides into this process. Chlamydiae are obligate intracellular “minimal” bacteria and causal agents of human and animal diseases with major economic and public health concern. For a long time the host range of chlamydiae has been underestimated. Recently, we discovered Chlamydia-related bacteria in great apes from Central Africa identified our closest relatives as novel hosts paving the way to understand the evolutionary adaption of chlamydiae to humans [1].

Several fascinating aspects distinguish the chlamydiae from other eubacteria. The most striking one is their extraordinary biphasic developmental cycle. In general, during the life cycle, chlamydiae alternate between elementary body (EB) and reticulate body (RB). The extracellular EBs (0,1 µm in size) are responsible for the dissemination of the infection whereas the RBs are important for replication. After attachment to an eukaryotic cells EBs get internalized and remain in special vacuoles termed inclusions. Within the inclusion EBs differentiate into the intracellular RBs (1 µm in size). These undergo repeated cycles of binary fission before they differentiate back to EBs and are released to start a new infection cycle [2]. Under stressful conditions, chlamydiae can enter a non-infectious but viable state, known as persistence. In vitro persistence is characterized by aberrant bodies (enlarged, non-dividing reticulate bodies), which are not capable of differentiating into EBs and remain associated with their host over long periods of time. Several stressors, including interferon-γ-induced tryptophan deprivation, iron chelation, and exposure to antimicrobial agents can induce chlamydial persistence [3], [4]. After removal of the stressor chlamydia can turn back into their normal cell cycle. Because of their intracellular lifestyle they do not need a cell wall (peptidoglycan) to resist osmotic challenges and for more than 20 years the structure could not been detected. However, despite the apparently missing cell wall, these pathogens are unexpectedly sensitive to cell wall biosynthesis inhibitors which is known as “Chlamydial anomaly”. In 2014, Maurelli and coworkes revealed evidence for the presence of circularly shaped peptidoglycan-like structures in chlamydiae [5].

The goal of this work is to gain more insights into the “Chlamydial anomaly” on a cellular and molecular level. For this reason a fluorescence-microscopy based system has been established to analyze cellular effects of cell wall targeting antibiotics on the chlamydial life cycle in a cell culture model. Specially the treatment with different beta-lactams, beside penicillin, and beta-lactamase inhibitors was investigated. The tested compounds resulted in varying numbers and morphological types of persisting cells in these cell wall lacking bacteria. For a better understanding of this phenomena proteins of the peptidoglycan biosynthesis pathway were investigated on a molecular level. In particular cell wall hydrolases like AmiA, the homolog of a hydrolase important for cytokinesis in E. coli. AmiA from Chlamydia pneumoniae was functionally analyzed using complementation experiments in the heterologous host E. coli as well as biochemical assays with the recombinant enzyme. The chlamydial AmiA restores the function and separates daughter cells in an E. coli amidase mutant. Interestingly, purified AmiA accepted both polymeric peptidoglycan as well as monomeric lipid II (the cell wall precursor) as a substrate. Noteworthy, AmiA showed an unexpected novel dual activity acting as amidase and penicillin-sensitive carboxypeptidase [6]. Functional conservation of AmiA implicates a role of this enzyme in chlamydial cell division. Additionally, the processing of lipid II caused by AmiA might subvert Nod2 receptor-mediated host response to muropeptides, contributing to long-term residence of chlamydiae in the host. However, a better understanding of the chlamydial lipid II processing and cell division machineries is needed to gain further insight into host response modulation and impact of persistent chlamydiae in patients. Further insights into the molecular mechanism on the basic of these findings could help to develop new treatment strategies against active and persistent chlamydial infections.

Note: A. Klöckner and C. Otten contributed equally.


References

1.
Klöckner A, Nagel M, Greub G, Aeby S, Hoffmann K, Liégeois F, Rouet F, De Benedetti S, Borel N, Henrichfreise B. Chlamydia-Related Bacteria in Free-Living and Captive Great Apes, Gabon. Emerg Infect Dis. 2016 Dec;22(12):2199-201. DOI: 10.3201/eid2212.150893 Externer Link
2.
Bedson SP, Bland JOW. A Morphological Study of Psittacosis Virus, with the Description of a Developmental Cycle. Br J Exp Pathol. 1932 Oct;13(5):461-6. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2048359/ Externer Link
3.
Hogan RJ, Mathews SA, Mukhopadhyay S, Summersgill JT, Timms P. Chlamydial persistence: beyond the biphasic paradigm. Infect Immun. 2004 Apr;72(4):1843-55. DOI: 10.1128/IAI.72.4.1843-1855.2004 Externer Link
4.
Borel N, Pospischil A, Hudson AP, Rupp J, Schoborg RV. The Role of Viable but Non-Infectious Developmental Forms in Chlamydial Biology. Front Cell Infect Microbiol. 2014;4(7):97. DOI: 10.3389/fcimb.2014.00097 Externer Link
5.
Liechti GW, Kuru E, Hall E, Kalinda A, Brun YV, VanNieuwenhze M, Maurelli AT. A new metabolic cell-wall labelling method reveals peptidoglycan in Chlamydia trachomatis. Nature. 2014 Feb 27;506(7489):507-10. DOI: 10.1038/nature12892 Externer Link
6.
Klöckner A, Otten C, Derouaux A, Vollmer W, Bühl H, De Benedetti S, Münch D, Josten M, Mölleken K, Sahl HG, Henrichfreise B. AmiA is a penicillin target enzyme with dual activity in the intracellular pathogen Chlamydia pneumoniae. Nat Commun. 2014 Jun 23;5:4201. DOI: 10.1038/ncomms5201 Externer Link