1,511
Views
3
CrossRef citations to date
0
Altmetric
Editorial - Commissioned

Legionella pneumophila-induced cell death: Two hosts, two responses

Pages 17-19 | Received 20 Sep 2017, Accepted 20 Sep 2017, Published online: 10 Nov 2017
This article refers to:
Differential expression of virulence genes in Legionella pneumophila growing in Acanthamoeba and human monocytes

Legionella pneumophila is a Gram-negative bacterium responsible of Legionnaire's disease, a severe form of pneumonia. L. pneumophila resides within natural and man-made aquatic systems. It shares these habitats with protozoa such as free-living amoebae that feed on bacteria. After uptake by amoeba, L. pneumophila is able to resist intracellular digestion and to multiply within this environmental host. Amoebae are considered as training ground for pathogenic bacteria such as L. pneumophila. Importantly, entry and intracellular replication of L. pneumophila within amoebae and mammalian macrophages display several similarities.Citation1 Once engulfed, L. pneumophila avoids fusion of the phagosome with lysosomes and creates a favorable environment for replication, the replicative vacuole, which is surrounded by the endoplasmic reticulum and mitochondria.Citation2 The ability of L. pneumophila to manipulate host cell functions is conferred by hundreds of effectors that are secreted or injected into the cytosol and the vacuole through type II (T2SS) and type IV (T4SS) secretion systems.Citation3,4 Intracellular growth of L. pneumophila increases its resistance to antimicrobials facilitating dispersion of the bacterium.Citation5

Although a number of mechanisms to infect amoebae and macrophages present common features and a shared evolutionary origin, some specificities were reported. For example, certain genetic loci allow L. pneumophila to infect macrophages, but not Acanthamoeba.Citation6 In contrast, L. pneumophila mutants that are defective in inhibiting host translation have lowered growth in the amoeba Dictyostelium but show no replication defect in macrophages.Citation7 Highlighting specific interactions between L. pneumophila and its different hosts is essential to understand the adaptation of Legionella to its multiple and evolutionarily distant hosts.

Another important issue is bacteria-induced cell death, which is relatively straightforward to observe, but difficult to characterize in detail. Difficulties come from the number of cell death pathways described to date. Bacteria could indeed induce cell death by activating apoptosis, pyroptosis, oncosis, necroptosis, NETosis, paraptosis or autophagic cell death.Citation8-10 In amoebae, the absence of certain families of proteins such as caspases, renders the classification of the bacterial-induced host cell death arduous. In addition, bacteria can possess an arsenal of different effectors that either activate or repress the host cell death.

In this issue of Virulence, Mou and Leung address the expression of L. pneumophila genes that are involved in human monocyte and Acanthamoeba cell death.Citation11 They also investigate the correlation of specific L. pneumophila gene expression patterns with the type of host cell death induced by the bacterium. The authors selected four set of genes, two which are involved in pyroptosis (flaA and sdhA) and two involved in apoptosis (vipD and sidF) of macrophages. The genes flaA and vipD encode respectively L. pneumophila flagellin and a phospholipase, and have been described to trigger host cell death.Citation12,13 In contrast, sdhA and sidF are T4SS-translocated effectors that inhibit cell death.Citation14,15

Mou and Leung point out differences in infection mechanisms depending on the host infected. Regarding genes related to pyroptosis, the authors observed a decrease of flaA expression and an increase of sdhA during infection of THP1 monocytes. Interestingly, the opposite result was obtained with the amoeba Acanthamoeba castellanii. Expression of flaA increased, while sdhA expression decreased. Expression of apoptosis-related genes vipD and sidF also differed during infections of both THP1 cells and A. castellanii. Infection of THP1 cells induced a decrease in mRNA levels of both vipD and sidF. In contrast, A. castellanii infection induced a very weak regulation of sidF and an earlier down-regulation of vipD, followed by a later up-regulation. Although the two genes involved in pyroptosis were differently expressed in L. pneumophila infecting THP1 and A. castellanii cells, they could promote a same phenomenon: repression of pyroptosis in monocytes or induction of cell death in amoebae. The biological relevance regarding the expression of apoptosis-related genes was less obvious, underlying the need to increase the panel of genes to study.

On the host side, the authors observed differences in uptake and replication of L. pneumophila. They found a higher replication of L. pneumophila in A. castellanii compared to THP1 cells. Moreover, the infection of monocytes with L. pneumophila was associated with a reduction of caspase-1 expression compared to uninfected cells. However, relationship between caspases expression and host cell death is not clear and need to be investigated. Overall, this study cumulated evidence suggesting that L. pneumophila could inhibit pyroptosis of THP1 cells.

The authors faced several limitations when they compared L. pneumophila infection of the two different hosts. For example, there is no evidence of the presence of caspases in amoebae, although caspase activities have been reported.Citation16 Instead, caspase-like proteins (metacaspase or paracaspase) have been discovered.Citation17 Despite similarities between caspases and caspase-like proteins, there are a few significant differences, such as the target cleavage site sequence.Citation17 Another issue is that, in addition to the programmed cell death, metacaspases contribute to several cellular functions. Thus, the metacaspase-1 is involved in encystation of A. castellanii.Citation18 Mou and Leung found that, over the incubation time, L. pneumophila induced an increasing metacaspase-1 expression, which was associated with the formation of cysts. This result has to be confronted with other studies showing that A. castellanii infected with L. pneumophila do not exhibit a cell wall containing cellulose as observed in mature cystCitation19 and present a low expression of metacaspase-1 at 48h post-infection.Citation20

In summary, Mou and Leung demonstrated that expression of L. pneumophila genes involved in host cell death is diametrically opposite depending on the infected host. This study contributes to a better understanding of L. pneumophila adaptation to its very large spectrum of hosts. The expression pattern of pyroptosis-related genes in the environmental amoebal host could suggest the need of host cell death to ensure dissemination of L. pneumophila. In contrast, the control of monocyte cell death could be essential to maintain infection, as Humans are accidental dead-end hosts.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

References

  • Escoll P, Rolando M, Gomez-Valero L, Buchrieser C. From Amoeba to Macrophages: Exploring the Molecular Mechanisms of Legionella pneumophila Infection in Both Hosts. Cur. Top Microbiol. Immunol. 2013;376:1–34. PMID:23949285
  • Isberg RR, O'Connor TJ, Heidtman M. The Legionella pneumophila replication vacuole: making a cosy niche inside host cells. Nat Rev Microbiol. 2009;7:13–24. doi:10.1038/nrmicro1967. PMID:19011659
  • Hubber A, Roy CR. Modulation of host cell function by Legionella pneumophila type IV effectors. Annu Rev Cell Dev Biol. 2010;26:261–83. doi:10.1146/annurev-cellbio-100109-104034. PMID:20929312
  • Truchan HK, Christman HD, White RC, Rutledge NS, Cianciotto NP. Type II Secretion Substrates of Legionella pneumophila Translocate Out of the Pathogen-Occupied Vacuole via a Semipermeable Membrane. MBio. 2017;8:e00870–17. https://doi.org/10.1128/mBio.00870-17.
  • Barker J, Scaife H, Brown MR. Intraphagocytic growth induces an antibiotic-resistant phenotype of Legionella pneumophila. Antimicrob Agents Chemother. 1995;39:2684–8. doi:10.1128/AAC.39.12.2684. PMID:8593002
  • Gao LY, Harb OS, Kwaik YA. Identification of macrophage-specific infectivity loci (mil) of Legionella pneumophila that are not required for infectivity of protozoa. Infect and Immun. 1998;66:883–92. PMID:9488371
  • Fontana MF, Banga S, Barry KC, Shen X, Tan Y, Luo ZQ, Vance RE. Secreted bacterial effectors that inhibit host protein synthesis are critical for induction of the innate immune response to virulent Legionella pneumophila. PLoS Pathogen. 2011;7:e1001289. doi:10.1371/journal.ppat.1001289. PMID:21390206
  • Jorgensen I, Rayamajhi M, Miao EA. Programmed cell death as a defence against infection. Nat Rev Immunol. 2017;17:151–64. doi:10.1038/nri.2016.147. PMID:28138137
  • Labbe K, Saleh M. Cell death in the host response to infection. Cell Death Differ. 2008;15:1339–49. doi:10.1038/cdd.2008.91. PMID:18566602
  • Mossine VV, Waters JK, Chance DL, Mawhinney TP. Transient Proteotoxicity of Bacterial Virulence Factor Pyocyanin in Renal Tubular Epithelial Cells Induces ER-Related Vacuolation and Can Be Efficiently Modulated by Iron Chelators. Toxico Sci: An offi J Soci Toxico. 2017;155:512. doi:10.1093/toxsci/kfw214. PMID:28123001
  • Mou Q, Leung PHM. Differential expression of virulence genes in Legionella pneumophila growing in Acanthamoeba and human monocytes. Virulence. 2017; 10.1080/21505594.2017.1373925
  • Silveira TN, Zamboni DS. Pore formation triggered by Legionella spp. is an Nlrc4 inflammasome-dependent host cell response that precedes pyroptosis. Infect and Immun. 2010;78:1403–13.
  • Zhu W, Hammad LA, Hsu F, Mao Y, Luo ZQ. Induction of caspase 3 activation by multiple Legionella pneumophila Dot/Icm substrates. Cellul Microbiol. 2013;15:1783–95. PMID:23773455
  • Banga S, Gao P, Shen X, Fiscus V, Zong WX, Chen L, Luo ZQ. Legionella pneumophila inhibits macrophage apoptosis by targeting pro-death members of the Bcl2 protein family. Proc Natl Acad Sci U S A. 2007;104:5121–6. doi:10.1073/pnas.0611030104. PMID:17360363
  • Ge J, Gong YN, Xu Y, Shao F. Preventing bacterial DNA release and absent in melanoma 2 inflammasome activation by a Legionella effector functioning in membrane trafficking. Proc Natl Acad Sci U S A. 2012;109:6193–8. doi:10.1073/pnas.1117490109. PMID:22474394
  • Wu D, Qiao K, Feng M, Fu Y, Cai J, Deng Y, Tachibana H, Cheng X. Apoptosis of Acanthamoeba castellanii Trophozoites Induced by Oleic Acid. J Eukaryot Microbiol. 2017. doi:10.1111/jeu.12454. [Epub ahead of print]
  • Saheb E, Trzyna W, Bush J. Caspase-like proteins: Acanthamoeba castellanii metacaspase and Dictyostelium discoideum paracaspase, what are their functions? J Biosci. 2014;39:909–16. doi:10.1007/s12038-014-9486-0. PMID:25431419
  • Trzyna WC, Legras XD, Cordingley JS. A type-1 metacaspase from Acanthamoeba castellanii. Microbiol Res. 2008;163:414–23. doi:10.1016/j.micres.2006.06.017. PMID:16891103
  • Mengue L, Regnacq M, Aucher W, Portier E, Hechard Y, Samba-Louaka A. Legionella pneumophila prevents proliferation of its natural host Acanthamoeba castellanii. Sci Rep. 2016;6:36448. doi:10.1038/srep36448. PMID:27805070
  • Ohno A, Kato N, Sakamoto R, Kimura S, Yamaguchi K. Temperature-dependent parasitic relationship between Legionella pneumophila and a free-living amoeba (Acanthamoeba castellanii). Appl Environ Microbiol. 2008;74:4585–8. doi:10.1128/AEM.00083-08. PMID:18502936