Next Article in Journal
Genome Sequence of a Thermoacidophilic Methanotroph Belonging to the Verrucomicrobiota Phylum from Geothermal Hot Springs in Yellowstone National Park: A Metagenomic Assembly and Reconstruction
Next Article in Special Issue
SARS-CoV-2–Legionella Co-Infections: A Systematic Review and Meta-Analysis (2020–2021)
Previous Article in Journal
Long-Term Effects of Soil Remediation with Willow Short Rotation Coppice on Biogeographic Pattern of Microbial Functional Genes
Previous Article in Special Issue
Water Age Effects on the Occurrence and Concentration of Legionella Species in the Distribution System, Premise Plumbing, and the Cooling Towers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Knowledge to Predict Pathogens: Legionella pneumophila Lifecycle Systematic Review Part II Growth within and Egress from a Host Cell

1
School of Nursing, Health, Exercise Science, The College of New Jersey, P.O. Box 7718, 2000 Pennington Rd., Ewing, NJ 08628, USA
2
Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH 43210, USA
3
Sustainability Institute, The Ohio State University, Columbus, OH 43210, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2022, 10(1), 141; https://doi.org/10.3390/microorganisms10010141
Submission received: 13 December 2021 / Revised: 30 December 2021 / Accepted: 4 January 2022 / Published: 11 January 2022
(This article belongs to the Special Issue Legionella pneumophila: A Microorganism with a Thousand Faces)

Abstract

:
Legionella pneumophila (L. pneumophila) is a pathogenic bacterium of increasing concern, due to its ability to cause a severe pneumonia, Legionnaires’ Disease (LD), and the challenges in controlling the bacteria within premise plumbing systems. L. pneumophila can thrive within the biofilm of premise plumbing systems, utilizing protozoan hosts for protection from environmental stressors and to increase its growth rate, which increases the bacteria’s infectivity to human host cells. Typical disinfectant techniques have proven to be inadequate in controlling L. pneumophila in the premise plumbing system, exposing users to LD risks. As the bacteria have limited infectivity to human macrophages without replicating within a host protozoan cell, the replication within, and egress from, a protozoan host cell is an integral part of the bacteria’s lifecycle. While there is a great deal of information regarding how L. pneumophila interacts with protozoa, the ability to use this data in a model to attempt to predict a concentration of L. pneumophila in a water system is not known. This systematic review summarizes the information in the literature regarding L. pneumophila’s growth within and egress from the host cell, summarizes the genes which affect these processes, and calculates how oxidative stress can downregulate those genes.

1. Introduction

1.1. Gap in the Literature

Due to the prevalence of L. pneumophila in the water system and the seriousness of legionellosis, a set of two respiratory diseases: Legionnaire’s Disease (LD) and the less severe Pontiac Fever, there has been a focus on L. pneumophila in the literature. Review articles separately discuss health effects, pathology, treatment, transmission, etiology, epidemiology, and infectivity [1,2,3,4,5,6,7,8,9]. A review article that combines these knowledge goals within the context of modeling the lifecycle of L. pneumophila in the water system to forecast concentration is needed. As environmental stress affects the regulation and processing of bacterial genes, it is an integral part in modeling the lifecycle of a pathogen. Data and knowledge pertinent to the impact of environmental stress on a pathogen’s lifecycle must be reviewed to assess their impact on the postulated model framework and the accuracy of a predictive model. In this, a convergence of the above-referenced fields must be reviewed and assessed for use within such a model. A review of this nature does not, to date, exist in the literature.

1.2. Need for This Review

This review seeks to summarize the information pertinent to developing a predictive growth model which incorporates the host–cell interactions of L. pneumophila and how environmental stressors affect these interactions. The lifecycle of L. pneumophila, as it pertains to the host cells involves three processes: uptake, growth, and egression. Considering the complexity of these processes, we have divided this systematic literature review into two parts, the invasion of the host cell [10], and the growth within and egress from the host cell, presented here. This paper focuses on the pathways, processes, and data for forecasting growth and egress of L. pneumophila within and from the host cell. Understanding how L. pneumophila interacts with host cells in its environment, particularly in biofilms, is vital in understanding how to treat L. pneumophila in the water system. Thus far, attempts to eradicate the bacteria from the water system have been made in vain [11,12]. However, understanding the genetics in which L. pneumophila can utilize host cells in its environment for protection and increased pathogenesis, and how certain environmental stressors can affect those genes is vital in understanding how to control L. pneumophila in the water system.
The mechanisms between L. pneumophila’s growth and egress in a protozoa and a human macrophage are similar, and the genetic knockdowns which prevent the bacteria from growing or egressing in a protozoan typically prevent growth and egress within a macrophage as well [13]. Additionally, if L. pneumophila is unable to replicate within and egress from a host cell in the environment, the bacteria will be less, or even potentially not at all, infectious to human macrophages [14]. Determining if chlorination or other environmental stressors of the water treatment process influences or causes, genetic knockdowns, which will decrease L. pneumophila’s ability to survive and thrive, is a first step in controlling legionellosis outbreaks. This review summarizes knowledge and data needed for a clearer picture of how environmental quality can govern growth within and egress from a host cell. A new means of controlling infectivity may rely on reduced growth or egress from host cells. These data are a vital initial foray into modeling these processes. This study provides said data from the literature which can be used in a predictive model.

1.3. General Growth Requirements

L. pneumophila thrives in fresh, warm water environments, with supportive nutrients and protozoan hosts [15]. Their ideal temperature range is 20–45 °C, with the bacteria being dormant below 20 °C and typically unable to survive at temperatures higher than 60 °C [16]. Legionella has been shown to survive 70 °C treatments for up to 30 min [17]. In premise plumbing systems, L. pneumophila utilizes the biofilm for protection against disinfectants and for access to nutrients and host cells, typically amoeba. Biofilms have been shown to be protective against environmental stressors, such as chlorine, to the bacteria [18,19]. The organic matter of the biofilm increases chlorine demand, which maximizes on the surface of the biofilm [20].
The rate in which chlorine penetrates the biofilm is dependent on the density of the biofilm, with older, more established biofilms being denser and therefore more resistant to chlorine [21]. In dense biofilms, as little as 10% of the bulk chlorine will penetrate the biofilm [21]. Growth of L. pneumophila in the biofilm is supported by high nutrient levels of iron, organic carbon, nitrogen, and phosphorous [22]. L. pneumophila has multiple iron acquisition pathways and is unable to grow in the absence of iron [23,24]. The bacteria produce siderophores to aid in iron acquisition in low iron environments, but are more robust in the high iron environment typical of a biofilm [25,26].

1.4. Antibiotic Resistance in L. pneumophila

Antibiotic resistance is of concern for any pathogen, particularly those associated with hospital acquired infections (HAIs). As such, the susceptibility of L. pneumophila to antibiotics has been studied since shortly after the discovery of the pathogen [27,28]. While there has been an increase in antibiotic resistance of L. pneumophila in environmental samples and patient cultures, there has yet to be a challenge in treating LD infections [29,30]. Research indicates that some antibiotic resistance may be due to environmental conditions and the relationship of the pathogen with host cells [31,32]. There is always the concern that antibiotic strains of L. pneumophila will become more virulent and more common place, as seen in the methicillin-resistant Staphylococcus aureus (MRSA) [33]. For that reason, it may be prudent to test for antibiotic resistance of L. pneumophila in premise plumbing samples, particularly in high risk buildings, such as hospitals [34].

1.5. Relationship with Host Cells

L. pneumophila is a facultative intracellular bacterium which can utilize host cells, such as amoeba, within the biofilm for further protection and to serve essentially as a unicellular incubator [35]. L. pneumophila grown intracellularly are smaller, more highly mobile, and have increased levels of microbial resistance when compared to bacterial progeny not grown in a host cell [31]. Acanthamoeba polyphaga (A. polyphaga) cysts are protective to L. pneumophila with chlorine concentrations as high as 50 mg/L, whereas free-living L. pneumophila is susceptible to chlorine concentrations as low as 2 mg/L [36,37].

1.6. Implications of This Study

The bacteria can replicate freely within in the host cell and can do so in multiple vacuoles and in counts typically over two dozen bacterium per vacuole [38]. Vacuoles can be expelled from the host cell prior to cell death [39]. The bacteria have been shown to be more infective to mammalian macrophages after having replicated within an amoeba host [35]. It is even theorized that the bacteria will not cause disease in humans unless it has replicated within a protozoan host first [40,41]. Therefore, bacteria growing and egressing from a host cell is a crucial part of L. pneumophila’s lifecycle in concern to human infectivity. However, current growth models for L. pneumophila in premise plumbing system do not incorporate the interactions with the host cell and the effects of disinfectants on the genes that dictate those interactions. This review is pertinent in summarizing the information needed to build said models.

2. Materials and Methods

2.1. Search Strategy

To determine the mechanisms, proteins, and genes involved in the growth of Legionella within and egress from a host cell, an exhaustive literature review was conducted from inception to January 2019. Google Scholar, PubMed, Scopus, Web of Science, Bioline International, and PLOS ONE were searched using the terms: “((Legionella) OR (Legionella pneumophila) OR (L. pneumophila) OR (Legionella longbeachae) OR (L. longbeachae) OR (Legionella bozemanii) OR (L. bozemanii)) AND ((Growth) OR (Intracellular Growth) OR (Replication) OR (Reproduction) OR (dot/icm) OR (Genetic Knockout) OR (Genetic Knockdown) OR (Genes for growth) OR (Host interaction) OR (Disinfection) OR (Acanthamoeba) OR (Acanthamoeba polyphaga) OR (Acanthamoeba castellanii) or (A. polyphaga) OR (A. castellanii) OR (Premise Plumbing) OR (Biofilms) OR (Egress) OR (Exit) OR (Mediated death) OR (Mediated exit) OR (Apoptosis)”. Relevant citations were forward and reverse searched, and imported into a Zotero library.

2.2. Eligibility Criteria

The inclusion criteria were Legionella studies which looked at the bacteria’s behavior, protein function, genetic function, genetic expression, or genetic change while growing within or egressing from a host cell. Studies had to be peer-reviewed and written in English.
Studies were excluded if they were performed in mice, focused on lifecycle stages outside of growth or egress, focused on free Legionella or cells outside of a host cell, were abstracts from conference proceedings, were letters to the editor, or were not refereed.

2.3. Study Selection

Over 2000 papers were imported to a Zotero library to review for relevance. Zotero’s automation was used to remove duplicate items. Titles and abstracts were reviewed for relevance by two reviewers working independently, then sorted by lifecycle stages, protein, or gene function. Studies were then determined to be eligible for modeling purposes if they were (1) performed in triplicate (2) utilized microbial techniques which only accounted for growth of bacteria within, or undergoing egress from, the host cell, while excluding additional bacterial uptake in the host cell (3) accounted for time and (4) able to be combined with similar data from the literature for one working model. Thirty-two studies evaluating proteins and twenty studies evaluating gene function contributing to the growth or egress of Legionella in a host cell were included in this review. Six studies were included as viable options to model growth rate efficiency within the cell and four studies were included to model egress rate efficiency. To be included in the modeling studies, a change in growth or egression rates based on a genetic knockdown needed to be measured. Nine studies overlapped in these categories for a total of 53 included studies. The Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines were used for this review. The 2020 PRISMA checklist can be found in Supplemental Material S1.

3. Results

3.1. Lifecycle and Legionella Containing Vacuoles

L. pneumophila has multiple ways of invading the host cell, resulting in the bacteria becoming encased in the phagosomal membrane [10]. The phagosomal membrane will alter its thickness to resemble the thinner endoplasmic reticulum (ER) vesicle, and invade the rough endoplasmic reticulum (RER) about 6 h after infection [42]. The bacteria are then able to intercept vesicular traffic from the ER exit sites to create an organelle that permits intracellular replication, preventing the destruction of the host cell—the Legionella containing vacuole (LCV) [43]. In the first hour, the mitochondria cluster about the LCV. Approximately 4–8 h after phagocytosis, the host cell’s ribosomes appear on the cytoplasmic side of the vacuolar membrane while L. pneumophila multiplies in the vacuole. At the end of the eighth hour, nearly all of the LCVs are studded with ribosomes and the bacteria have a doubling time of about 2 h [44,45]. L. pneumophila will remain within the LCV until hundreds of bacteria fill the vacuole and the monocyte ruptures [46].
L. pneumophila utilizes the Dot/Icm Type IV Secretion System (T4SS) and the Type II Secretion System (TS2), in order to evade the phagosome-lysosome binding process, allowing the bacteria to replicate within the host cell [46,47,48]. These systems share several components, most likely having a common evolutionary origin [49,50,51]. Together, they export over 300 effector proteins. TS2 exports multiple effectors which contribute to the broad host range of L. pneumophila TS2, including protesase, RNase, lipase, phospholipase A, phospholipase C, lysophospholipase A, and tartrate-sensitive and tartrate-resistant acid phosphatase [52,53]. While the bacteria are replicating within the host cell, the LCV recruits host proteins in order to aid in LCV maintenance, including calnexin, Sec22b, BiP, SAR1, and Rab1, which are host factors involved in the endoplasmic reticulum (ER) recruitment process [54,55,56,57]. Some of these proteins are soluble in the cytosol and enter the LCV. They aide the LCV in avoiding intracellular degradation by establishing an ER-associated replicative compartment [54,58,59]. Individual effector proteins and host cell proteins will be discussed in further detail in Table 1.
LCVs that were expelled from host cells were found to have as many as 104 L. pneumophila within a single vesicle [60]. More than 90% of vesicles containing viable L. pneumophila cells, expelled from A. polyphaga or A. castellanii, were of a respirable size, <5 µm [39]. It is theorized that one is more likely to become infected with L. pneumophila after inhaling a contaminated vesicle as opposed to free bacteria [61].
Table 1. Proteins associated with L. pneumophila’s ability to grow within and egress from a host cell.
Table 1. Proteins associated with L. pneumophila’s ability to grow within and egress from a host cell.
ProteinFunctionTargetReference
AnkBTranslocated effector, allows proliferation of bacteria.LCV[47,62]
AnkGPrevents apoptosis of host cell allowing for continued replication of L. pneumophila in mammalian hosts. Host protein gCq1R(p32)[63]
DsbA2Catalyzes the disulfide bond formation required for the extracytoplasmic assembly of the T4SS system of L. pneumophila. Dot/Icm T4SS[64]
Dot/Icm Type 4 Secretion System (T4SS)Translocates over 300 proteins into the host cells. Modulates host processes including phagosome-lysosome binding, promotion of ubiquitin conjugates, and suppression of dendritic cell formation. Icm/Dot Translocated substrates (IDTS) [65,66,67,68]
DrrARequired for host cytotoxicity. Recruits and activates Rab1 on the plasma membrane-derived organelles. Host vesicular transport[56]
IcmSWMediates a conformational change facilitating T4SS recognition of the effector protein, thereby enhancing effector protein deliveryTranslocation domain in the effector protein[69]
IcmQForms pores in lipid membranes by utilizing a chaperone/substrate relationship. [70]
IcmRBinds to the N-terminal of IcmQ inhibiting membrane insertion and pore formation. IcmQ[70]
LbtPSidephore transport protein which allows for growth in iron-limiting conditions. Prevents premature exit of macrophage due to low nutrients. [26]
LbtUSidephore transport protein which imports iron-bound legiobactin. Alos for growth in iron-limiting conditions.Legiobactin[26]
LegC3Inhibits SNARE and Rab GTPase dependent membrane fusion pathway [71]
LegK1Modulates macrophage defense and inflammatory response during infection of a host cell.NF-kB[72]
LegK2Efficient recruitment of endoplasmic reticulum markers allowing for timely intracellular replication and ER uptake of the LCV. T4SS[73]
LtpDIntracellular bacterial replication. Phosphatidylinositol 3-phosphate[74]
PieAAvoids phagososome/lysosome binding. Allows for growth in the cell. LCV[75,76,77]
PI4PLocalization of effectors to LCV early during infection. [48,78]
PmrAAllows for intracellular growth in host cells. Dot/Icm Type 4 secretion system[79]
RalFExchange factor for the ARF family of GTPase. Required for the localization of ARF of LCV ARF [80]
Rap1Allows for intracellular bacterial replication. [81]
RpkALocalizes endosomal membranes, specifically recruited to the phagosome.LCV[13]
RpoSStimulates intracellular replication and osmotic resistance. Growth phase stress resistance in protozoa. Maximum flagellin expression. fliA, flaA, mip[82,83,84]
SidCInvolved in recruiting host ER proteins to the surface of the LCV, allowing for intracellular bacterial replication. LCV, PtdIns(4)P[85]
SidFAllows for more bacterial replication by making host cell resistant to apoptosis. NIP3, Mcl-rambo[86]
SidHImportant in early phase of infection. Inhibits cells death. [87]
SetAAllows for bacterial virulence in the post-exponential growth phase by preventing entry of the LCBB into the endocytic network. LCV[88]

3.2. Flagella

Similar to the various pathways L. pneumophila can use to infect host cells, the bacteria utilize flagellin differently within different hosts and during various stages of development. Flagellin and motility are required for the colonization of A. castellanii to activate the NLRC4 pathway, but have an adverse effect during bacterial replication in mammalian lungs [89]. L. pneumophila require flagellin to induce apoptosis of the host cell, but flagellin are not required for replication within the host cell [90]. The flagella initiates the caspase 9 and effector caspase 3, activating the pro-apoptotic protein Bax and inhibiting the anti-apoptotic protein X-linked inhibitor of apoptosis (XIAP) via the inhibition of the Akt pathway [90]. Macrophages and dendritic cells use flagellin to assess the virulence of bacteria [91]. Flagellin is responsible for activating the nuclear factor κB (NF-κB), p38 mitogen-activated protein kinases (MAPK), Jun N-terminal kinase (JNK), and transforming growth factor β-associated kinase 1 (TAK1), which induce interleukin-8 (IL-8) activation, the human immune response to L. pneumophila infection [92]. Nod-like receptors NOD1 and NOD2 are partially responsible for neutrophil recruitment and cytokine production in the mammalian lung [93,94,95]. Flagellin can be translocated by the Dot/Icm complex into the host cell cytosol, where macrophages and dendritic cells can use the protein to assess the virulence of the bacteria [91,96]. Although L. pneumophila cannot replicate within dendritic cells, it still utilizes the Dot/Icm complex to establish ER-derived LCV within the cell [97]. The interaction between the flagellin and the host cell Nod-like receptors, Ipaf and NLRC4, which both activate caspase 1 or TLR5, can induce the expression of pro-inflammatory cytokines [89,98,99]. NLRC4 is not localized to a distinct structure within the cell, allowing inflammasomes to gain access to different subsets of substrates. It also activates caspase 7, which promotes non-apoptotic functions such as LCV maturation and bacterial degradation [100].

3.3. Interferons

Type I interferons (IFN-α/β), which boost the immune system in response to an infection, are induced by L. pneumophila after the Dot/Icm complex translocates bacterial DNA into the cytosol of the host cell [101,102,103]. A downstream signaling adaptor in the stimulator of interferon genes (STING) pathway is required for type I IFN induction in response to an upstream sensor of the cytosolic DNA [104,105,106]. Type I IFNs directs the activation of both Stat 1 homodimers and IFN-stimulated gene factor 3 (ISGF-3) which are integral in the activation of the IFN-I autocrine loop [107]. Type II IFNs (IFN-γ) utilize the classical pathway to activate macrophages with only the Stat1 homodimer. Both Type I and Type II IFNs play an integral role in the innate immune response of the macrophage to intracellular microbes [107]. Even low doses of IFN are effective in preventing the replication of L. pneumophila in macrophage host cells [108]. IFN-activated macrophages inhibit the bacteria from proliferating mainly by reactive oxygen intermediate and reactive nitrogen intermediate independent mechanisms, and secondarily by nutritionally dependent mechanisms [109]. One key protein in suppressing the IFN response to L. pneumophila, SdhA, is discussed in Table 2 [110].

3.4. Mediated Cell Death

Naip5, a Nod-like receptor protein, initiates cell death through the activation of caspase 1, causing a pore formation and resulting in pyroptosis, caspase 1-mediated cell death [123,124,125,126,127]. Caspase 1 activation is mediated in response to a translocated Dot/Icm substrate and recombinant flagellin in the cytosol, and will not occur in IPAF mutant cells [100,123,128]. This process stimulates autophagy in macrophages, resulting in the redirected trafficking of the LCV to lysosomes [129,130].
Despite normal caspase 1 function, if the cytosolic protein NAIP5 is defective, the macrophage is permissive to the bacteria replicating [127,131]. All macrophages require a competent Dot/Icm complex in order for the cell to undergo apoptosis [132]. ASC, the apoptosis-associated speck-like protein, containing a caspase recruitment domain, is an adaptor protein not associated with pyroptosis, but rather aids in mediated-cell death via an independent inflammasome pathway [123]. ASC plays a role in the negative regulation of caspase 1-dependent host cell death [133]. ASC is required for the secretion of inflammatory cytokines IL-1β and IL-18 [123,128,134]. Cytokine processing occurs in a single, large, punctate structure in host cells, where ASC and caspase 1 are recruited [134]. Caspase 1 is required for efficient cytokine processing, as a mutant form of caspase 1 is unable to support cytokine cleavage [134].
Caspase 3 is essential for apoptosis in monocytes, macrophages, and alveolar epithelial cells. While the bacteria activate caspase 3 upon invasion, apoptotic death is not executed until late stages of infection, after the bacteria have completed replication [135]. The protein is associated with effective Dot/Icm mediated anti-apoptotic stimuli which cause the cell to resist the apoptotic inducer during bacterial replication [100]. The caspase proteins and non-apoptotic functions of executioner caspases are modulated, temporally and spatially, during infection, determining permissiveness to intracellular bacterial proliferation [100].
The host death of infected macrophages occurs in a biphasic model. The induction of apoptosis occurs during the early stages of infection and the independent and temporal induction of necrosis occurs during the late stages of intracellular replication [121]. In a similar manner as the bacteria kills the protozoan host, necrosis and cytolysis of macrophages by L. pneumophila is mediated by pore-forming activity or toxin [121,136]. The pore-forming activity is signaled after the bacteria have finished replicating in the host cell. Mutants which are incapable of pore-forming activity can replicate within the host cell but are unable to lyse the host cell and egress. They will eventually be released by the host, most likely by apoptotic death [121].
L. pneumophila utilizes multiple mechanisms to induce cell death, which can vary depending on the host cell. L. pneumophila uses type II and IV secretion systems to cleave large subunit ribosomal RNA resulting in decreased mitochondrial messenger RNAs in Dictyostelium discoideum (D. discoideum) [137]. However, even in other protozoan hosts, such as A. castellanii, L. pneumophila do not use this method. Contact-dependent cytotoxicity is required for the egress of L. pneumophila from the amoeba, while it is not needed for the bacteria to survive and thrive within the host cell [113]. In dendritic cells, mitochondria-regulated apoptosis occurs within 1 h [138]. This process is initiated by caspase 3 or BH3-only proteins [96].

3.5. Stress

Amoeba are known as a Trojan Horse for pathogenic microorganisms, serving as both reservoirs and vehicles for the bacteria in the environment [14]. Furthermore, amoeba can serve as a unicellular incubator, allowing the bacteria to adapt to life within a human macrophage, favoring pathogenesis [14]. Environmental stressors such as high temperatures, unfavorable pH, osmotic pressure, or presence of disinfectants can cause amoeba to encyst, a life-stage particularly protective to intracellular bacteria [61]. The double layered cyst is particularly resilient, surviving in temperatures from −20 °C to 42 °C and showing resistance against disinfectants, such as chlorine [139]. The amoeba will return to the trophozoic form when environmental conditions are more favorable. L. pneumophila have been observed in both trophozoites and cyst hosts [140]. Vesicles expelled from protozoan host cells have demonstrated resilience when exposed to biocides in cooling towers for up to 24 h, ultrasound, and vast temperature ranges (−70 °C to 35 °C) [39].

4. Discussion

4.1. L. pneumophila in Premise Plumbing Systems

Legionellosis is of particular concern in large premise plumbing systems, such as those found in hospitals or hotels, due to water stagnation, institutional hot water being kept under 50 °C, and disinfectant dissipating throughout the large systems [141,142,143,144]. The elderly and immunocompromised have higher incidence rates of LD, making hospitals and nursing care facilities of even greater concern [145,146,147]. Patients with ambulatory impediments may take longer to shower, leading to longer exposure times to L. pneumophila if it is present in the premise plumbing system [148]. Due to the seriousness of LD and the increasing incidences of legionellosis throughout the US, there have been many campaigns to eradicate the bacteria from premise plumbing systems, specifically in biofilms where the bacteria are significantly more difficult to treat than free living L. pneumophila. The endosymbiotic relationship L. pneumophila has with protozoa makes eradication of the bacteria within the biofilm of premise plumbing systems exceedingly difficult [149]. As L. pneumophila use the protozoa as a reservoir the bacteria are particularly resistant to typical disinfection measures [135]. L. pneumophila’s lifecycle and factors affecting the bacteria’s persistence and virulence have been well documented. However, a comprehensive literature review providing information necessary to model L. pneumophila’s growth within and egress from host cells within a biofilm was not available, necessitating this article.

4.2. Genetic Knockdowns

Genetic knockdowns or mutations would not only affect the individual bacterium, but extend to its progeny, affecting all future generations. Therefore, a knockdown or mutation that is nonlethal, but lowers the bacterium’s infectivity would then lower the infectivity of all of the bacterium’s descendants. Genetic knockdown information, which affects L. pneumophila’s ability to replicate within and egress from the host cell, was drawn from the literature. Data are only reported in this article if they were sufficient to use in a mechanistic model, requiring that: (a) they were reported in a peer-reviewed journal, (b) they had greater than three data points, (c) they were validated, (d) the genes effected are responsible for pathogenesis, and (e) they were comparable to the other data provided. Table 2 summarizes the functions of genes essential for L. pneumophila to grow within and egress from host cells. Variations in methodologies, timeframes, and units reported (i.e., log vs. percentage reduction) present a challenge in using data from the literature in one comprehensive model. In this article, the rates at which the modified bacteria had reduced capacity to grow within or egress from the host cell after the knockdown when compared to the wildtype are reported in the same units as the cited article in the “change in growth efficiency” or the “change in egress rate efficiency” columns in Table 3 and Table 4, respectively. The degradation of growth or egress rates are also expressed as percentages in Table 3 and Table 4, for the sake of uniformity in modeling. Percentages were chosen over log reductions as some genetic knockdowns cause only a minor change in growth or egress. For modeling purposes, it is recommended that future research report their results in percentage reductions. There is a great deal of redundancies within the gene functions in L. pneumophila, making the bacteria more robust to environmental stressors and adaptable to its environmental conditions. However, it is clear that specific genetic knockdowns, such as the ones described in Table 3 and Table 4, reduce the bacteria’s virulence in the biofilm and premise plumbing systems.

4.3. Contribution to the Literature

This study summarizes information from the literature, which is vital to model the growth within and egress from host cells, and how these mechanisms can be influenced by environmental stressors, such as disinfectants. In recent years the importance of replication within host cells for the virulence of L. pneumophila in human macrophages has become clear [35]. It is important to incorporate this knowledge into future predictive models so as to fully understand the infectivity of the bacteria to humans. This paper summarizes the information available in the literature to allow for said more substantial predictive models.

4.4. Limitation of This Systematic Review

This review serves to summarize the data available in the literature regarding how oxidative stress affects L. pneumophila’s lifecycle, specifically during its replication and egress phases. It looks at how oxidative stress affects genetic regulation in the bacteria and how those genes affect these lifecycle traits. However, there is still much work to be done in understanding the lifecycle of L. pneumophila, its interaction with the host cell, how environmental stressors change this relationship, and what genes are involved. Furthermore, this study did not focus on the antibiotic resistance of L. pneumophila in the environment or in humans. This will undoubtedly be a topic of high importance in the future and would be helpful to incorporate in monitoring for the bacteria and future risk analysis.

5. Future Directions

This review summarizes knowledge and data that can be used to build an intracellular growth model for L. pneumophila in the biofilm. It is clear that replication within a host cell is vital to incorporating infectivity and virulence while modeling the lifecycle of L. pneumophila. The genes that regulate protein secretion and ultimately the phylogenic characteristics of effective replication in and egress from the host cells are used to model the lifecycle of the bacteria. The stochastic method was used in modeling this data as a result of the uncertainty and variability of environmental stressors effects, as well as the lack of data conducive to modeling. Stochastic methods allow for systems and data uncertainty and variability to be accounted for and used in the model estimates. The modeling framework for the intracellular growth part of the predictive model is represented in Figure 1. The environmental quality and oxidative stress impact genetic knockdown which in turn affects phylogenetic outcomes, resulting in replication rate degradation. The modeling framework in Figure 1 allows for a mechanistic model of replication rates due to oxidative stress.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms10010141/s1, Supplementary Material S1: PRISMA 2020 Checklist.

Author Contributions

A.L.M. and M.H.W. conceived and designed concepts for this paper; A.L.M. and M.H.W. performed the literature review and research; A.L.M. wrote the paper. All authors have read and agreed to the published version of the manuscript.

Funding

Funding for this research is partially supported by The Quantitative Microbial Risk Assessment Interdisciplinary Instructional Institute supported by the National Institute of General Medical Sciences of the National Institutes of Health (NIH) under Award Number R25GM108593.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data used in this study are included in the published article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Carrington, C.B. Pathology of Legionnaires’ Disease. Ann. Intern. Med. 1979, 90, 496–499. [Google Scholar] [CrossRef] [PubMed]
  2. Balows, A.; Fraser, D.W. International Symposium on Legionnaires’ Disease, 13–15 November 1978, Atlanta, Georgia. Ann. Intern. Med. 1979, 90, 489–714. [Google Scholar]
  3. Edelstein, P.H.; Meyer, R.D. Legionnaires’ Disease. Chest 1984, 85, 114–120. [Google Scholar] [CrossRef] [PubMed]
  4. Swartz, M.N. Clinical Aspects of Legionnaires’ Disease. Ann. Intern. Med. 1979, 90, 492–495. [Google Scholar] [CrossRef]
  5. Isenberg, H.D. Microbiology of Legionnaires’ Disease Bacterium. Ann. Intern. Med. 1979, 90, 499. [Google Scholar] [CrossRef] [PubMed]
  6. Rose, J.B. Future Health Assessment and Risk-Management Integration for Infectious Diseases and Biological Weapons for Deployed US Forces. In Strategies to Protect the Health of Deployed US Forces: Assessing Health Risks to Deployed US Forces: Workshop Proceedings; National Academies Press: Washington, DC, USA, 2000; pp. 59–112. [Google Scholar]
  7. Eickhoff, T.C. Epidemiology of Legionnaires’ Disease. Ann. Intern. Med. 1979, 90, 499. [Google Scholar] [CrossRef] [PubMed]
  8. Muder, R.R.; Victor, L.Y.; Woo, A.H. Mode of Transmission of Legionella pneumophila: A Critical Review. Arch. Intern. Med. 1986, 146, 1607–1612. [Google Scholar] [CrossRef]
  9. Craun, G.F.; Brunkard, J.M.; Yoder, J.S.; Roberts, V.A.; Carpenter, J.; Wade, T.; Calderon, R.L.; Roberts, J.M.; Beach, M.J.; Roy, S.L. Causes of Outbreaks Associated with Drinking Water in the United States from 1971 to 2006. Available online: http://cmr.asm.org (accessed on 16 February 2018).
  10. Mraz, A.L.; Weir, M.H. Knowledge to Predict Pathogens: Legionella pneumophila Lifecycle Critical Review Part I Uptake into Host Cells. Water 2018, 10, 132. [Google Scholar] [CrossRef] [Green Version]
  11. Kim, B.R.; Anderson, J.E.; Mueller, S.A.; Gaines, W.A.; Kendall, A.M. Literature Review—Efficacy of Various Disinfectants against Legionella in Water Systems. Water Res. 2002, 36, 4433–4444. [Google Scholar] [CrossRef]
  12. Atlas, R.M. Legionella: From Environmental Habitats to Disease Pathology, Detection and Control. Environ. Microbiol. 1999, 1, 283–293. [Google Scholar] [CrossRef]
  13. Riyahi, T.Y.; Frese, F.; Steinert, M.; Omosigho, N.N.; Glöckner, G.; Eichinger, L.; Orabi, B.; Williams, R.S.B.; Noegel, A.A. RpkA, a Highly Conserved GPCR with a Lipid Kinase Domain, Has a Role in Phagocytosis and Anti-Bacterial Defense. PLoS ONE 2011, 6, e27311. [Google Scholar] [CrossRef] [PubMed]
  14. Barker, J.; Brown, M. Trojan-Horses of the Microbial World—Protozoa and the Survival of Bacterial Pathogens in the Environment. Microbiology 1994, 140, 1253–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Fliermans, C.B.; Cherry, W.B.; Orrison, L.H.; Smith, S.J.; Tison, D.L.; Pope, D.H. Ecological Distribution of Legionella pneumophila. Appl. Environ. Microbiol. 1981, 41, 9–16. [Google Scholar] [CrossRef] [Green Version]
  16. Health and Safety Executive. Legionnaires’ Disease: The Control of Legionella Bacteria in Water Systems; Health and Safety Executive: Bootle, UK, 2013. [Google Scholar]
  17. Farhat, M.; Moletta-Denat, M.; Frère, J.; Onillon, S.; Trouilhé, M.-C.; Robine, E. Effects of Disinfection on Legionella Spp., Eukarya, and Biofilms in a Hot Water System. Appl. Environ. Microbiol. 2012, 78, 6850–6858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Cargill, K.L.; Pyle, B.H.; Sauer, R.L.; McFeters, G.A. Effects of Culture Conditions and Biofilm Formation on the Iodine Succeptibility of Legionella pneumophila. Can. J. Microbiol. 1992, 38, 423–429. [Google Scholar] [CrossRef] [PubMed]
  19. Cooper, I.R.; Hanlon, G.W. Resistance of Legionella pneumophila Serotype 1 Biofilms to Chlorine-Based Disinfection. J. Hosp. Infect. 2010, 74, 152–159. [Google Scholar] [CrossRef]
  20. Butterfield, P.W.; Camper, A.K.; Biederman, J.A.; Bargmeyer, A.M. Minimizing Biofilm in the Presence of Iron Oxides and Humic Substances. Water Res. 2002, 36, 3898–3910. [Google Scholar] [CrossRef]
  21. Chen, X.; Stewart, P.S. Chlorine Penetration into Artificial Biofilm Is Limited by a Reaction-Diffusion Interaction. Environ. Sci. Technol. 1996, 30, 2078–2083. [Google Scholar] [CrossRef]
  22. Jjemba, P.K.; Weinrich, L.A.; Cheng, W.; Giraldo, E.; Lechevallier, M.W. Regrowth of Potential Opportunistic Pathogens and Algae in Reclaimed-Water Distribution Systems. Appl. Environ. Microbiol. 2010, 76, 4169–4178. [Google Scholar] [CrossRef] [Green Version]
  23. Cianciotto, N.P. Iron Acquisition by Legionella pneumophila. BioMetals 2007, 20, 323–331. [Google Scholar] [CrossRef]
  24. Orsi, N. The Antimicrobial Activity of Lactoferrin: Current Status and Perspectives. BioMetals 2004, 17, 189–196. [Google Scholar] [CrossRef]
  25. Naylor, J.; Cianciotto, N.P. Cytochrome c Maturation Proteins Are Critical for in Vivo Growth of Legionella pneumophila. FEMS Microbiol. Lett. 2004, 241, 249–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. O’Connor, T.J.; Zheng, H.; VanRheenen, S.M.; Ghosh, S.; Cianciotto, N.P.; Isberg, R.R. Iron Limitation Triggers Early Egress by the Intracellular Bacterial Pathogen Legionella pneumophila. Infect. Immun. 2016, 84, 2185–2197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Marre, R.; Medeiros, A.A.; Pasculle, A.W. Characterization of the Beta-Lactamases of Six Species of Legionella. J. Bacteriol. 1982, 151, 216–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Clarke, C.W.; Marlin, G.E. Antibiotic Treatment of Pneumonia. Aust. N. Z. J. Med. 1982, 12, 85–89. [Google Scholar] [CrossRef] [PubMed]
  29. Jia, X.; Ren, H.; Nie, X.; Li, Y.; Li, J.; Qin, T. Antibiotic Resistance and Azithromycin Resistance Mechanism of Legionella pneumophila Serogroup 1 in China. Antimicrob. Agents Chemother. 2019, 63, e00768-19. [Google Scholar] [CrossRef]
  30. Pappa, O.; Chochlakis, D.; Sandalakis, V.; Dioli, C.; Psaroulaki, A.; Mavridou, A. Antibiotic Resistance of Legionella pneumophila in Clinical and Water Isolates—A Systematic Review. Int. J. Environ. Res. Public. Health 2020, 17, 5809. [Google Scholar] [CrossRef] [PubMed]
  31. Barker, J.; Scaife, H.; Brown, M.R. Intraphagocytic Growth Induces an Antibiotic-Resistant Phenotype of Legionella pneumophila. Antimicrob. Agents Chemother. 1995, 39, 2684–2688. [Google Scholar] [CrossRef] [Green Version]
  32. Li, L.; Mendis, N.; Trigui, H.; Faucher, S.P. Transcriptomic Changes of Legionella pneumophila in Water. BMC Genom. 2015, 16, 637. [Google Scholar] [CrossRef] [PubMed]
  33. Enright, M.C.; Robinson, D.A.; Randle, G.; Feil, E.J.; Grundmann, H.; Spratt, B.G. The Evolutionary History of Methicillin-Resistant Staphylococcus Aureus (MRSA). Proc. Natl. Acad. Sci. USA 2002, 99, 7687–7692. [Google Scholar] [CrossRef] [Green Version]
  34. De Giglio, O.; Napoli, C.; Lovero, G.; Diella, G.; Rutigliano, S.; Caggiano, G.; Montagna, M.T. Antibiotic Susceptibility of Legionella pneumophila Strains Isolated from Hospital Water Systems in Southern Italy. Environ. Res. 2015, 142, 586–590. [Google Scholar] [CrossRef] [Green Version]
  35. Harb, O.S.; Gao, L.-Y.; Kwaik, Y.A. From Protozoa to Mammalian Cells: A New Paradigm in the Life Cycle of Intracellular Bacterial Pathogens. Environ. Microbiol. 2000, 2, 251–265. [Google Scholar] [CrossRef] [PubMed]
  36. Kilvington, S.; Price, J. Survival of Legionella pneumophila within Cysts of Acanthamoeba Polyphaga Following Chlorine Exposure. J. Appl. Bacteriol. 1990, 68, 519–525. [Google Scholar] [CrossRef]
  37. Miyamoto, H.; Ogawa, M.; Maruta, K.; Yamamoto, C.; Taniguchi, H.; Yoshida, S.-I.; Nikaido, Y. Temperature Effects on Legionella pneumophila Killing by and Multiplication in Phagocytes of Guinea Pigs. Microbiol. Immunol. 1995, 39, 647–654. [Google Scholar] [CrossRef]
  38. Steinert, M.; Birkness, K.; White, E.; Fields, B.; Quinn, F. Mycobacterium Avium Bacilli Grow Saprozoically in Coculture with Acanthamoeba Polyphaga and Survive within Cyst Walls. Appl. Environ. Microbiol. 1998, 64, 2256–2261. [Google Scholar] [CrossRef] [Green Version]
  39. Berk, S.G.; Ting, R.S.; Turner, G.W.; Ashburn, R.J. Production of Respirable Vesicles Containing Live Legionella pneumophila Cells by Two Acanthamoeba Spp. Appl. Environ. Microbiol. 1998, 64, 279–286. [Google Scholar] [CrossRef] [Green Version]
  40. Horwitz, M.A.; Silverstein, S.C. Legionnaires’ Disease Bacterium (Legionella pneumophila) Multiplies Intracellularly in Human Monocytes. J. Clin. Investig. 1980, 66, 441–450. [Google Scholar] [CrossRef] [Green Version]
  41. Nash, T.W.; Libby, D.M.; Horwitz, M.A. Interaction between the Legionnaires’ Disease Bacterium (Legionella pneumophila) and Human Alveolar Macrophages. Influence of Antibody, Lymphokines, and Hydrocortisone. J. Clin. Investig. 1984, 74, 771–782. [Google Scholar] [CrossRef] [Green Version]
  42. Tilney, L.G.; Harb, O.S.; Connelly, P.S.; Robinson, C.G.; Roy, C.R. How the Parasitic Bacterium Legionella pneumophila Modifies Its Phagosome and Transforms It into Rough ER: Implications for Conversion of Plasma Membrane to the ER Membrane. J. Cell Sci. 2001, 114, 4637–4650. [Google Scholar] [CrossRef] [PubMed]
  43. Kagan, J.C.; Roy, C.R. Legionella Phagosomes Intercept Vesicular Traffic from Endoplasmic Reticulum Exit Sites. Nat. Cell Biol. 2002, 4, 945–954. [Google Scholar] [CrossRef] [PubMed]
  44. Horwitz, M.A. Formation of a Novel Phagosome by the Legionnaires’ Disease Bacterium (Legionella pneumophila) in Human Monocytes. J. Exp. Med. 1983, 158, 1319–1331. [Google Scholar] [CrossRef] [Green Version]
  45. Steinert, M.; Hentschel, U.; Hacker, J. Legionella pneumophila: An Aquatic Microbe Goes Astray. FEMS Microbiol. Rev. 2002, 26, 149–162. [Google Scholar] [CrossRef] [PubMed]
  46. Horwitz, M.A. The Legionnaires’ Disease Bacterium (Legionella pneumophila) Inhibits Phagosome-Lysosome Fusion in Human Monocytes. J. Exp. Med. 1983, 158, 2108–2126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Price, C.T.D.; Kwaik, Y.A. The Transcriptome of Legionella pneumophila-Infected Human Monocyte-Derived Macrophages. PLoS ONE 2014, 9, e114914. [Google Scholar] [CrossRef]
  48. Hubber, A.; Arasaki, K.; Nakatsu, F.; Hardiman, C.; Lambright, D.; De Camilli, P.; Nagai, H.; Roy, C.R. The Machinery at Endoplasmic Reticulum-Plasma Membrane Contact Sites Contributes to Spatial Regulation of Multiple Legionella Effector Proteins. PLoS Pathog. 2014, 10, e1004222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Peabody, C.R.; Chung, Y.J.; Yen, M.-R.; Vidal-Ingigliardi, D.; Pugsley, A.P.; Saier, M.H. Type II Protein Secretion and Its Relationship to Bacterial Type IV Pili and Archaeal Flagella. Microbiology 2003, 149, 3051–3072. [Google Scholar] [CrossRef]
  50. Nunn, D. Bacterial Type II Protein Export and Pilus Biogenesis: More than Just Homologies? Trends Cell Biol. 1999, 9, 402–408. [Google Scholar] [CrossRef]
  51. Thanassi, D.G.; Hultgren, S.J. Multiple Pathways Allow Protein Secretion across the Bacterial Outer Membrane. Curr. Opin. Cell Biol. 2000, 12, 420–430. [Google Scholar] [CrossRef]
  52. Tyson, J.Y.; Pearce, M.M.; Vargas, P.; Bagchi, S.; Mulhern, B.J.; Cianciotto, N.P. Multiple Legionella pneumophila Type II Secretion Substrates, Including a Novel Protein, Contribute to Differential Infection of the Amoebae Acanthamoeba Castellanii, Hartmannella Vermiformis, and Naegleria Lovaniensis. Infect. Immun. 2013, 81, 1399–1410. [Google Scholar] [CrossRef] [Green Version]
  53. Rossier, O.; Starkenburg, S.R.; Cianciotto, N.P. Legionella pneumophila Type II Protein Secretion Promotes Virulence in the A/J Mouse Model of Legionnaires’ Disease Pneumonia. Infect. Immun. 2004, 72, 310–321. [Google Scholar] [CrossRef] [Green Version]
  54. Derré, I.; Isberg, R.R. Legionella pneumophila Replication Vacuole Formation Involves Rapid Recruitment of Proteins of the Early Secretory System. Infect. Immun. 2004, 72, 3048–3053. [Google Scholar] [CrossRef] [Green Version]
  55. Kagan, J.C.; Stein, M.-P.; Pypaert, M.; Roy, C.R. Legionella Subvert the Functions of Rab1 and Sec22b to Create a Replicative Organelle. J. Exp. Med. 2004, 199, 1201–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Murata, T.; Delprato, A.; Ingmundson, A.; Toomre, D.K.; Lambright, D.G.; Roy, C.R. The Legionella pneumophila Effector Protein DrrA Is a Rab1 Guanine Nucleotide-Exchange Factor. Nat. Cell Biol. 2006, 8, 971–977. [Google Scholar] [CrossRef]
  57. Swanson, M.S.; Isberg, R.R. Association of Legionella pneumophila with the Macrophage Endoplasmic Reticulum. Infect. Immun. 1995, 63, 3609–3620. [Google Scholar] [CrossRef] [Green Version]
  58. Derré, I.; Isberg, R.R. LidA, a Translocated Substrate of the Legionella pneumophila Type IV Secretion System, Interferes with the Early Secretory Pathway. Infect. Immun. 2005, 73, 4370–4380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Robinson, C.G.; Roy, C.R. Attachment and Fusion of Endoplasmic Reticulum with Vacuoles Containing Legionella pneumophila. Cell. Microbiol. 2006, 8, 793–805. [Google Scholar] [CrossRef]
  60. Rowbotham, T.J. Current Views on the Relationships between Amoebae, Legionellae and Man. Isr. J. Med. Sci. 1986, 22, 678–689. [Google Scholar]
  61. Greub, G.; Raoult, D. Microorganisms Resistant to Free-Living Amoebae. Clin. Microbiol. Rev. 2004, 17, 413–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Al-Khodor, S.; Price, C.T.; Habyarimana, F.; Kalia, A.; Abu Kwaik, Y. A Dot/Icm-Translocated Ankyrin Protein of Legionella pneumophila Is Required for Intracellular Proliferation within Human Macrophages and Protozoa. Mol. Microbiol. 2008, 70, 908–923. [Google Scholar] [CrossRef] [Green Version]
  63. Lührmann, A.; Nogueira, C.V.; Carey, K.L.; Roy, C.R. Inhibition of Pathogen-Induced Apoptosis by a Coxiella Burnetii Type IV Effector Protein. Proc. Natl. Acad. Sci. USA 2010, 107, 18997–19001. [Google Scholar] [CrossRef] [Green Version]
  64. Kpadeh, Z.Z.; Jameson-Lee, M.; Yeh, A.J.; Chertihin, O.; Shumilin, I.A.; Dey, R.; Day, S.R.; Hoffman, P.S. Disulfide Bond Oxidoreductase DsbA2 of Legionella pneumophila Exhibits Protein Disulfide Isomerase Activity. J. Bacteriol. 2013, 195, 1825–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Meir, A.; Chetrit, D.; Liu, L.; Roy, C.R.; Waksman, G. Legionella DotM Structure Reveals a Role in Effector Recruiting to the Type 4B Secretion System. Nat. Commun. 2018, 9, 507. [Google Scholar] [CrossRef] [Green Version]
  66. Schroeder, G.N. The Toolbox for Uncovering the Functions OfLegionellaDot/Icm Type IVb Secretion System Effectors: Current State and Future Directions. Front. Cell. Infect. Microbiol. 2017, 7, 528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Harding, C.R.; Stoneham, C.A.; Schuelein, R.; Newton, H.; Oates, C.V.; Hartland, E.L.; Schroeder, G.N.; Frankel, G. The Dot/Icm Effector SdhA Is Necessary for Virulence of Legionella pneumophila in Galleria Mellonella and A/J Mice. Infect. Immun. 2013, 81, 2598–2605. [Google Scholar] [CrossRef] [Green Version]
  68. Ivanov, S.S.; Roy, C.R. Modulation of Ubiquitin Dynamics and Suppression of DALIS Formation by the Legionella pneumophila Dot/Icm System. Cell. Microbiol. 2009, 11, 261–278. [Google Scholar] [CrossRef] [Green Version]
  69. Cambronne, E.D.; Roy, C.R. The Legionella pneumophila IcmSW Complex Interacts with Multiple Dot/Icm Effectors to Facilitate Type IV Translocation. PLoS Pathog. 2007, 3, e188. [Google Scholar] [CrossRef] [Green Version]
  70. Duménil, G.; Montminy, T.P.; Tang, M.; Isberg, R.R. IcmR-Regulated Membrane Insertion and Efflux by the Legionella pneumophila IcmQ Protein. J. Biol. Chem. 2004, 279, 4686–4695. [Google Scholar] [CrossRef] [Green Version]
  71. Bennett, T.L.; Kraft, S.M.; Reaves, B.J.; Mima, J.; O’Brien, K.M.; Starai, V.J. LegC3, an Effector Protein from Legionella pneumophila, Inhibits Homotypic Yeast Vacuole Fusion In Vivo and In Vitro. PLoS ONE 2013, 8, e56798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ge, J.; Xu, H.; Li, T.; Zhou, Y.; Zhang, Z.; Li, S.; Liu, L.; Shao, F. A Legionella Type IV Effector Activates the NF-KappaB Pathway by Phosphorylating the IkappaB Family of Inhibitors. Proc. Natl. Acad. Sci. USA 2009, 106, 13725–13730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hervet, E.; Charpentier, X.; Vianney, A.; Lazzaroni, J.-C.; Gilbert, C.; Atlan, D.; Doublet, P. Protein Kinase LegK2 Is a Type IV Secretion System Effector Involved in Endoplasmic Reticulum Recruitment and Intracellular Replication of Legionella pneumophila. Infect. Immun. 2011, 79, 1936–1950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Harding, C.R.; Mattheis, C.; Mousnier, A.; Oates, C.V.; Hartland, E.L.; Frankel, G.; Schroeder, G.N. LtpD Is a Novel Legionella pneumophila Effector That Binds Phosphatidylinositol 3-Phosphate and Inositol Monophosphatase IMPA1. Infect. Immun. 2013, 81, 4261–4270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Brand, B.C.; Sadosky, A.B.; Shuman, H.A. The Legionella pneumophila Icm Locus: A Set of Genes Required for Intracellular Multiplication in Human Macrophages. Mol. Microbiol. 1994, 14, 797–808. [Google Scholar] [CrossRef]
  76. Vogel, J.P.; Andrews, H.L.; Wong, S.K.; Isberg, R.R. Conjugative Transfer by the Virulence System of Legionella pneumophila. Science 1998, 279, 873–876. [Google Scholar] [CrossRef] [PubMed]
  77. Berger, K.H.; Merriam, J.J.; Isberg, R.R. Altered Intracellular Targeting Properties Associated with Mutations in the Legionella pneumophila DotA Gene. Mol. Microbiol. 1994, 14, 809–822. [Google Scholar] [CrossRef] [PubMed]
  78. Ninio, S.; Celli, J.; Roy, C.R. A Legionella pneumophila Effector Protein Encoded in a Region of Genomic Plasticity Binds to Dot/Icm-Modified Vacuoles. PLoS Pathog. 2009, 5, e1000278. [Google Scholar] [CrossRef] [Green Version]
  79. Zusman, T.; Aloni, G.; Halperin, E.; Kotzer, H.; Degtyar, E.; Feldman, M.; Segal, G. The Response Regulator PmrA Is a Major Regulator of the Icm/Dot Type IV Secretion System in Legionella pneumophila and Coxiella Burnetii. Mol. Microbiol. 2007, 63, 1508–1523. [Google Scholar] [CrossRef]
  80. Nagai, H.; D Cambronne, E.; Kagan, J.C.; Carlos Amor, J.; Kahn, R.; Roy, C. A C-Terminal Translocation Signal Required for Dot/Icm-Dependent Delivery of the Legionella RalF Protein to Host Cells. Proc. Natl. Acad. Sci. USA 2005, 102, 826–831. [Google Scholar] [CrossRef] [Green Version]
  81. Schmölders, J.; Manske, C.; Otto, A.; Hoffmann, C.; Steiner, B.; Welin, A.; Becher, D.; Hilbi, H. Comparative Proteomics of Purified Pathogen Vacuoles Correlates Intracellular Replication of Legionella pneumophila with the Small GTPase Ras-Related Protein 1 (Rap1). Mol. Cell. Proteomics 2017, 16, 622–641. [Google Scholar] [CrossRef] [Green Version]
  82. Molofsky, A.B.; Swanson, M.S. Differentiate to Thrive: Lessons from the Legionella pneumophila Life Cycle. Mol. Microbiol. 2004, 53, 29–40. [Google Scholar] [CrossRef] [Green Version]
  83. Hales, L.M.; Shuman, H.A. The Legionella pneumophila RpoS Gene Is Required for Growth within Acanthamoeba Castellanii. J. Bacteriol. 1999, 181, 4879–4889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Bachman, M.A.; Swanson, M.S. Genetic Evidence That Legionella pneumophila RpoS Modulates Expression of the Transmission Phenotype in Both the Exponential Phase and the Stationary Phase. Infect. Immun. 2004, 72, 2468–2476. [Google Scholar] [CrossRef] [Green Version]
  85. Hsu, F.; Luo, X.; Qiu, J.; Teng, Y.-B.; Jin, J.; Smolka, M.B.; Luo, Z.-Q.; Mao, Y. The Legionella Effector SidC Defines a Unique Family of Ubiquitin Ligases Important for Bacterial Phagosomal Remodeling. Proc. Natl. Acad. Sci. USA 2014, 111, 10538–10543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Banga, S.; Gao, P.; Shen, X.; Fiscus, V.; Zong, W.-X.; Chen, L.; Luo, Z.-Q. Legionella pneumophila Inhibits Macrophage Apoptosis by Targeting Pro-Death Members of the Bcl2 Protein Family. Proc. Natl. Acad. Sci. USA 2007, 104, 5121–5126. [Google Scholar] [CrossRef] [Green Version]
  87. Luo, Z.-Q. Targeting One of Its Own: Expanding Roles of Substrates of the Legionella pneumophila Dot/Icm Type IV Secretion System. Front. Microbiol. 2011, 2, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Heidtman, M.; Chen, E.J.; Moy, M.-Y.; Isberg, R.R. Large-Scale Identification of Legionella pneumophila Dot/Icm Substrates That Modulate Host Cell Vesicle Trafficking Pathways. Cell. Microbiol. 2009, 11, 230–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Pereira, M.S.; Morgantetti, G.F.; Massis, L.M.; Horta, C.V.; Hori, J.I.; Zamboni, D.S. Activation of NLRC4 by Flagellated Bacteria Triggers Caspase-1–Dependent and–Independent Responses to Restrict Legionella pneumophila Replication in Macrophages and in Vivo. J. Immunol. 2011, 187, 6447–6455. [Google Scholar] [CrossRef] [Green Version]
  90. Takamatsu, R.; Takeshima, E.; Ishikawa, C.; Yamamoto, K.; Teruya, H.; Heuner, K.; Higa, F.; Fujita, J.; Mori, N. Inhibition of Akt/GSK3beta Signalling Pathway by Legionella pneumophila Is Involved in Induction of T-Cell Apoptosis. Biochem. J. 2010, 427, 57–67. [Google Scholar] [CrossRef]
  91. Miao, E.A.; Andersen-Nissen, E.; Warren, S.E.; Aderem, A. TLR5 and Ipaf: Dual Sensors of Bacterial Flagellin in the Innate Immune System. Semin. Immunopathol. 2007, 29, 275–288. [Google Scholar] [CrossRef]
  92. Takamatsu, R.; Teruya, H.; Takeshima, E.; Ishikawa, C.; Matsumoto, K.; Mukaida, N.; Li, J.-D.; Heuner, K.; Higa, F.; Fujita, J.; et al. Molecular Characterization of Legionella pneumophila-Induced Interleukin-8 Expression in T Cells. BMC Microbiol. 2010, 10, 1. [Google Scholar] [CrossRef] [Green Version]
  93. Archer, K.A.; Ader, F.; Kobayashi, K.S.; Flavell, R.A.; Roy, C.R. Cooperation between Multiple Microbial Pattern Recognition Systems Is Important for Host Protection against the Intracellular Pathogen Legionella pneumophila. Infect. Immun. 2010, 78, 2477–2487. [Google Scholar] [CrossRef] [Green Version]
  94. Berrington, W.R.; Iyer, R.; Wells, R.D.; Smith, K.D.; Skerrett, S.J.; Hawn, T.R. NOD1 and NOD2 Regulation of Pulmonary Innate Immunity to Legionella pneumophila. Eur. J. Immunol. 2010, 40, 3519–3527. [Google Scholar] [CrossRef]
  95. Frutuoso, M.S.; Hori, J.I.; Pereira, M.S.F.; Junior, D.S.L.; Sônego, F.; Kobayashi, K.S.; Flavell, R.A.; Cunha, F.Q.; Zamboni, D.S. The Pattern Recognition Receptors Nod1 and Nod2 Account for Neutrophil Recruitment to the Lungs of Mice Infected with Legionella pneumophila. Microbes Infect. 2010, 12, 819–827. [Google Scholar] [CrossRef] [PubMed]
  96. Shin, S. Innate Immunity to Intracellular Pathogens: Lessons Learned from Legionella pneumophila. Adv. Appl. Microbiol. 2012, 79, 43–71. [Google Scholar] [CrossRef]
  97. Neild, A.L.; Roy, C.R. Immunity to Vacuolar Pathogens: What Can We Learn from Legionella? Cell. Microbiol. 2004, 6, 1011–1018. [Google Scholar] [CrossRef]
  98. Spörri, R.; Joller, N.; Hilbi, H.; Oxenius, A. A Novel Role for Neutrophils As Critical Activators of NK Cells. J. Immunol. 2008, 181, 7121–7130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Shin, S.; Case, C.L.; Archer, K.A.; Nogueira, C.V.; Kobayashi, K.S.; Flavell, R.A.; Roy, C.R.; Zamboni, D.S. Type IV Secretion-Dependent Activation of Host MAP Kinases Induces an Increased Proinflammatory Cytokine Response to Legionella pneumophila. PLoS Pathog. 2008, 4, e1000220. [Google Scholar] [CrossRef]
  100. Amer, A.O. Modulation of Caspases and Their Non-Apoptotic Functions by Legionella pneumophila. Cell. Microbiol. 2010, 12, 140–147. [Google Scholar] [CrossRef] [PubMed]
  101. Luo, Z.-Q.; Isberg, R.R. Multiple Substrates of the Legionella pneumophila Dot/Icm System Identified by Interbacterial Protein Transfer. Proc. Natl. Acad. Sci. USA 2004, 101, 841–846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Stetson, D.B.; Medzhitov, R. Recognition of Cytosolic DNA Activates an IRF3-Dependent Innate Immune Response. Immunity 2006, 24, 93–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Opitz, B.; Vinzing, M.; Van, L.; Schmeck, B.; Heine, G.; Günther, S.; Preissner, R.; Slevogt, H.; N’Guessan, P.D.; Eitel, J.; et al. Legionella pneumophila Induces IFNβ in Lung Epithelial Cells via IPS-1 and IRF3, Which Also Control Bacterial Replication. J. Biol. Chem. 2006, 281, 36173–36179. [Google Scholar] [CrossRef] [Green Version]
  104. Ishikawa, H.; Barber, G.N. STING Is an Endoplasmic Reticulum Adaptor That Facilitates Innate Immune Signalling. Nature 2008, 455, 674–678. [Google Scholar] [CrossRef] [PubMed]
  105. Ishikawa, H.; Ma, Z.; Barber, G.N. STING Regulates Intracellular DNA-Mediated, Type i Interferon-Dependent Innate Immunity. Nature 2009, 461, 788–792. [Google Scholar] [CrossRef] [Green Version]
  106. Lippmann, J.; Müller, H.C.; Naujoks, J.; Tabeling, C.; Shin, S.; Witzenrath, M.; Hellwig, K.; Kirschning, C.J.; Taylor, G.A.; Barchet, W.; et al. Dissection of a Type I Interferon Pathway in Controlling Bacterial Intracellular Infection in Mice. Cell. Microbiol. 2011, 13, 1668–1682. [Google Scholar] [CrossRef] [Green Version]
  107. Plumlee, C.R.; Lee, C.; Beg, A.A.; Decker, T.; Shuman, H.A.; Schindler, C. Interferons Direct an Effective Innate Response to Legionella pneumophila Infection. J. Biol. Chem. 2009, 284, 30058–30066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Schiavoni, G.; Mauri, C.; Carlei, D.; Belardelli, F.; Pastoris, M.C.; Proietti, E. Type I IFN Protects Permissive Macrophages from Legionella pneumophila Infection through an IFN-Gamma-Independent Pathway. J. Immunol. Baltim. Md 1950 2004, 173, 1266–1275. [Google Scholar]
  109. Gebran, S.J.; Yamamoto, Y.; Newton, C.; Klein, T.W.; Friedman, H. Inhibition of Legionella pneumophila Growth by Gamma Interferon in Permissive A/J Mouse Macrophages: Role of Reactive Oxygen Species, Nitric Oxide, Tryptophan, and Iron(III). Infect. Immun. 1994, 62, 3197–3205. [Google Scholar] [CrossRef] [Green Version]
  110. Monroe, K.M.; McWhirter, S.M.; Vance, R.E. Identification of Host Cytosolic Sensors and Bacterial Factors Regulating the Type I Interferon Response to Legionella pneumophila. PLoS Pathog. 2009, 5, e1000665. [Google Scholar] [CrossRef]
  111. Marra, A.; Blander, S.J.; Horwitz, M.A.; Shuman, H.A. Identification of a Legionella pneumophila Locus Required for Intracellular Multiplication in Human Macrophages. Proc. Natl. Acad. Sci. USA 1992, 89, 9607–9611. [Google Scholar] [CrossRef] [Green Version]
  112. Segal, G.; Purcell, M.; Shuman, H.A. Host Cell Killing and Bacterial Conjugation Require Overlapping Sets of Genes within a 22-Kb Region of the Legionella pneumophila Genome. Proc. Natl. Acad. Sci. USA 1998, 95, 1669–1674. [Google Scholar] [CrossRef] [Green Version]
  113. Bandyopadhyay, P.; Byrne, B.; Chan, Y.; Swanson, M.S.; Steinman, H.M. Legionella pneumophila Catalase-Peroxidases Are Required for Proper Trafficking and Growth in Primary Macrophages. Infect. Immun. 2003, 71, 4526–4535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Roy, C.R.; Isberg, R.R. Topology of Legionella pneumophila DotA: An Inner Membrane Protein Required for Replication in Macrophages. Infect. Immun. 1997, 65, 571–578. [Google Scholar] [CrossRef] [Green Version]
  115. Liu, Y.; Luo, Z.-Q. The Legionella pneumophila Effector SidJ Is Required for Efficient Recruitment of Endoplasmic Reticulum Proteins to the Bacterial Phagosome. Infect. Immun. 2007, 75, 592–603. [Google Scholar] [CrossRef] [Green Version]
  116. Brassinga, A.K.C.; Kinchen, J.M.; Cupp, M.E.; Day, S.R.; Hoffman, P.S.; Sifri, C.D. Caenorhabditis Is a Metazoan Host for Legionella. Cell. Microbiol. 2010, 12, 343–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Roy, C.R.; Berger, K.H.; Isberg, R.R. Legionella pneumophila DotA Protein Is Required for Early Phagosome Trafficking Decisions That Occur within Minutes of Bacterial Uptake. Mol. Microbiol. 1998, 28, 663–674. [Google Scholar] [CrossRef] [Green Version]
  118. Furugen, M.; Higa, F.; Hibiya, K.; Teruya, H.; Akamine, M.; Haranaga, S.; Yara, S.; Koide, M.; Tateyama, M.; Mori, N.; et al. Legionella pneumophila Infection Induces Programmed Cell Death, Caspase Activation, and Release of High-Mobility Group Box 1 Protein in A549 Alveolar Epithelial Cells: Inhibition by Methyl Prednisolone. Respir. Res. 2008, 9, 39. [Google Scholar] [CrossRef] [Green Version]
  119. Bandyopadhyay, P.; Steinman, H.M. Catalase-Peroxidases of Legionella pneumophila: Cloning of the KatA Gene and Studies of KatA Function. J. Bacteriol. 2000, 182, 6679–6686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Bandyopadhyay, P.; Steinman, H.M. Legionella pneumophila Catalase-Peroxidases: Cloning of the KatB Gene and Studies of KatB Function. J. Bacteriol. 1998, 180, 5369–5374. [Google Scholar] [CrossRef] [Green Version]
  121. Alli, O.A.; Gao, L.Y.; Pedersen, L.L.; Zink, S.; Radulic, M.; Doric, M.; Abu Kwaik, Y. Temporal Pore Formation-Mediated Egress from Macrophages and Alveolar Epithelial Cells by Legionella pneumophila. Infect. Immun. 2000, 68, 6431–6440. [Google Scholar] [CrossRef] [PubMed]
  122. Laguna, R.K.; Creasey, E.A.; Li, Z.; Valtz, N.; Isberg, R.R. A Legionella pneumophila-Translocated Substrate That Is Required for Growth within Macrophages and Protection from Host Cell Death. Proc. Natl. Acad. Sci. USA 2006, 103, 18745–18750. [Google Scholar] [CrossRef] [Green Version]
  123. Case, C.L.; Shin, S.; Roy, C.R. Asc and Ipaf Inflammasomes Direct Distinct Pathways for Caspase-1 Activation in Response to Legionella pneumophila. Infect. Immun. 2009, 77, 1981–1991. [Google Scholar] [CrossRef] [Green Version]
  124. Derré, I.; Isberg, R.R. Macrophages from Mice with the Restrictive Lgn1 Allele Exhibit Multifactorial Resistance to Legionella pneumophila. Infect. Immun. 2004, 72, 6221–6229. [Google Scholar] [CrossRef] [Green Version]
  125. Silveira, T.N.; Zamboni, D.S. Pore Formation Triggered by Legionella Spp. Is an Nlrc4 Inflammasome-Dependent Host Cell Response That Precedes Pyroptosis. Infect. Immun. 2010, 78, 1403–1413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Whitfield, N.N.; Byrne, B.G.; Swanson, M.S. Mouse Macrophages Are Permissive to Motile Legionella Species That Fail To Trigger Pyroptosis. Infect. Immun. 2010, 78, 423–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Lamkanfi, M.; Amer, A.; Kanneganti, T.-D.; Muñoz-Planillo, R.; Chen, G.; Vandenabeele, P.; Fortier, A.; Gros, P.; Núñez, G. The Nod-like Receptor Family Member Naip5/Birc1e Restricts Legionella pneumophila Growth Independently of Caspase-1 Activation. J. Immunol. 2007, 178, 8022–8027. [Google Scholar] [CrossRef] [Green Version]
  128. Zamboni, D.S.; Kobayashi, K.S.; Kohlsdorf, T.; Ogura, Y.; Long, E.M.; Vance, R.E.; Kuida, K.; Mariathasan, S.; Dixit, V.M.; Flavell, R.A. The Birc1e Cytosolic Pattern-Recognition Receptor Contributes to the Detection and Control of Legionella pneumophila Infection. Nat. Immunol. 2006, 7, 318. [Google Scholar] [CrossRef] [PubMed]
  129. Amer, A.O.; Swanson, M.S. Autophagy Is an Immediate Macrophage Response to Legionella pneumophila. Cell. Microbiol. 2005, 7, 765–778. [Google Scholar] [CrossRef] [Green Version]
  130. Joshi, A.D.; Swanson, M.S. Secrets of a Successful Pathogen: Legionella Resistance to Progression Along the Autophagic Pathway. Front. Microbiol. 2011, 2, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Coers, J.; Vance, R.E.; Fontana, M.F.; Dietrich, W.F. Restriction of Legionella pneumophila Growth in Macrophages Requires the Concerted Action of Cytokine and Naip5/Ipaf Signalling Pathways. Cell. Microbiol. 2007, 9, 2344–2357. [Google Scholar] [CrossRef] [PubMed]
  132. Zink, S.D.; Pedersen, L.; Cianciotto, N.P.; Abu-Kwaik, Y. The Dot/Icm Type IV Secretion System of Legionella pneumophila Is Essential for the Induction of Apoptosis in Human Macrophages. Infect. Immun. 2002, 70, 1657–1663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Case, C.L.; Roy, C.R. Asc Modulates the Function of NLRC4 in Response to Infection of Macrophages by Legionella pneumophila. mBio 2011, 2, e00117-11. [Google Scholar] [CrossRef] [Green Version]
  134. Broz, P.; von Moltke, J.; Jones, J.W.; Vance, R.E.; Monack, D.M. Differential Requirement for Caspase-1 Autoproteolysis in Pathogen-Induced Cell Death and Cytokine Processing. Cell Host Microbe 2010, 8, 471–483. [Google Scholar] [CrossRef] [Green Version]
  135. Abu-Zant, A.; Santic, M.; Molmeret, M.; Jones, S.; Helbig, J.; Abu Kwaik, Y. Incomplete Activation of Macrophage Apoptosis during Intracellular Replication of Legionella pneumophila. Infect. Immun. 2005, 73, 5339–5349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Gao, L.-Y.; Abu Kwaik, Y. The Mechanism of Killing and Exiting the Protozoan Host Acanthamoeba Polyphaga by Legionella pneumophila. Environ. Microbiol. 2000, 2, 79–90. [Google Scholar] [CrossRef] [PubMed]
  137. Zhang, C.; Kuspa, A. Transcriptional Down-Regulation and RRNA Cleavage in Dictyostelium Discoideum Mitochondria during Legionella pneumophila Infection. PLoS ONE 2009, 4, e5706. [Google Scholar] [CrossRef] [Green Version]
  138. Nogueira, C.V.; Lindsten, T.; Jamieson, A.M.; Case, C.L.; Shin, S.; Thompson, C.B.; Roy, C.R. Rapid Pathogen-Induced Apoptosis: A Mechanism Used by Dendritic Cells to Limit Intracellular Replication of Legionella pneumophila. PLoS Pathog. 2009, 5, e1000478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Kahane, S.; Dvoskin, B.; Mathias, M.; Friedman, M.G. Infection of Acanthamoeba Polyphaga with Simkania Negevensis and S. Negevensis Survival within Amoebal Cysts. Appl. Environ. Microbiol. 2001, 67, 4789–4795. [Google Scholar] [CrossRef] [Green Version]
  140. Winiecka-Krusnell, J.; Linder, E. Bacterial Infections of Free-Living Amoebae. Res. Microbiol. 2001, 152, 613–619. [Google Scholar] [CrossRef]
  141. Blackburn, B.G.; Craun, G.F.; Yoder, J.S.; Hill, V.; Calderon, R.L.; Chen, N.; Lee, S.H.; Levy, D.A.; Beach, M.J. Surveillance for Waterborne-Disease Outbreaks Associated with Drinking Water--United States, 2001–2002. Morb. Mortal. Wkly. Rep. Surveill. Summ. Wash. DC 2002 2004, 53, 23–45. [Google Scholar]
  142. Kool, J.L.; Carpenter, J.C.; Fields, B.S. Effect of Monochloramine Disinfection of Municipal Drinking Water on Risk of Nosocomial Legionnaires’ Disease. Lancet Lond. Engl. 1999, 353, 272–277. [Google Scholar] [CrossRef]
  143. Barbaree, J.M.; Fields, B.S.; Feeley, J.C.; Gorman, G.W.; Martin, W.T. Isolation of Protozoa from Water Associated with a Legionellosis Outbreak and Demonstration of Intracellular Multiplication of Legionella pneumophila. Appl. Environ. Microbiol. 1986, 51, 422–424. [Google Scholar] [CrossRef] [Green Version]
  144. Ashbolt, N.J. Microbial Contamination of Drinking Water and Disease Outcomes in Developing Regions. Toxicology 2004, 198, 229–238. [Google Scholar] [CrossRef]
  145. Winn, W.C. Legionnaires Disease: Historical Perspective. Clin. Microbiol. Rev. 1988, 1, 60–81. [Google Scholar] [CrossRef]
  146. Legionella; Cianciotto, N.P.; Hacker, J.; Lück, P.C.; Fields, B.S.; Marre, R.; Frosch, M.; Abu Kwaik, Y.; Bartlett, C. American Society of Microbiology; 2002; ISBN 978-1-55581-230-0. Available online: https://www.ncbi.nlm.nih.gov/books/NBK215661/ (accessed on 12 December 2021).
  147. Dooling, K.L.; Toews, K.A.; Hicks, L.A.; Garrison, L.E.; Bachaus, B.; Zansky, S.; Carpenter, L.R.; Schaffner, B.; Parker, E.; Petit, S.; et al. Morbidity and Mortality Weekly Report. MMWR 2015, 64, 1190–1193. [Google Scholar]
  148. Wilkes, C.R.; Mason, A.D.; Hern, S.C. Probability Distributions for Showering and Bathing Water-Use Behavior for Various US Subpopulations. Risk Anal. Int. J. 2005, 25, 317–337. [Google Scholar] [CrossRef] [PubMed]
  149. National Research Council (US) Committee on Indicators for Waterborne Pathogens. Indicators for Water Pathogens; National Academies Press (US): Washington, DC, USA, 2004; Chapter 3 Ecology and Evolution of Waterborne Pathogens and Indicator Organism. [Google Scholar]
  150. Vincent, C.D.; Vogel, J.P. The Legionella pneumophila IcmS-LvgA Protein Complex Is Important for Dot/Icm-Dependent Intracellular Growth. Mol. Microbiol. 2006, 61, 596–613. [Google Scholar] [CrossRef]
  151. Jameson-Lee, M.; Garduño, R.A.; Hoffman, P.S. DsbA2 (27 KDa Com1-like Protein) of Legionella pneumophila Catalyses Extracytoplasmic Disulphide-Bond Formation in Proteins Including the Dot/Icm Type IV Secretion System. Mol. Microbiol. 2011, 80, 835–852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Coers, J.; Kagan, J.C.; Matthews, M.; Nagai, H.; Zuckman, D.M.; Roy, C.R. Identification of Icm Protein Complexes That Play Distinct Roles in the Biogenesis of an Organelle Permissive for Legionella pneumophila Intracellular Growth. Mol. Microbiol. 2000, 38, 719–736. [Google Scholar] [CrossRef] [PubMed]
  153. Molmeret, M.; Alli, O.A.; Radulic, M.; Susa, M.; Doric, M.; Kwaik, Y.A. The C-Terminus of IcmT Is Essential for Pore Formation and for Intracellular Trafficking of Legionella pneumophila within Acanthamoeba Polyphaga. Mol. Microbiol. 2002, 43, 1139–1150. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Modeling framework for intracellular growth of L. pneumophila in a host cell.
Figure 1. Modeling framework for intracellular growth of L. pneumophila in a host cell.
Microorganisms 10 00141 g001
Table 2. Genes associated with L. pneumophila’s ability to grow within and egress from a host cell.
Table 2. Genes associated with L. pneumophila’s ability to grow within and egress from a host cell.
GeneFunctionTargetReference
Dot/Icm ComplexAvoids phagososome/lysosome binding. Allows for growth in the cell and genetic conjugation. LCV[75,76,77,111,112]
ankBIntracellular replication in U937, A. polyphaga, and human monocyte-derived macrophages. [62]
ccm locusGrowth, intracellular infection and virulence, especially in low iron environmentsCytochrome c[25]
dotAIntracellular replication within macrophages, required in the immediate stages of infection to prevent lysozyme fusion. Not required for growth within amoeba or nematodes. [75,113,114,115,116,117]
dotBNot required for intracellular growth in amoeba. [113]
dotOInfection of A549 alveolar epithelial cells. Activates caspase 3, 8, 9, and 1. Released HMGB1.[118]
katABifunctional catalase-peroxidase. Keeps hydrogen peroxidase levels low in the cell allowing intracellular multiplication. LAMP-1, recruits phagosomes[113,119,120]
katBBifunctional catalase-peroxidase. Keeps hydrogen peroxidase levels low in the cell allowing for intracellular multiplication, LAMP-1, recruits phagosomes[113,119,120]
pilDIntracellular growth in U937 and amoeba Type II Secretion System[53]
ribExpression of pore-forming toxin/activity [121]
sidJGrowth in macrophage and amoeba [115]
sdjAGrowth in protozoan, but not macrophages [115]
sdhAPrevention of cell death-Mutation has increased nuclear degradation, mitochondrial distribution, membrane permeability, and caspase activationType I IFN expression[110,122]
sdeCEfficient intracellular growth [101]
Table 3. Effect of genetic knockdowns on the growth of L. pneumophila within a host cell.
Table 3. Effect of genetic knockdowns on the growth of L. pneumophila within a host cell.
GeneHost CellChange in Growth Rate Efficiency 1Degradation of Growth Rate 2Process
dotAU93735–56%35–56%Phagosome–lysosome fusion occurs [77].
dotAA. CastellanniIncapable of replication100%Phagosome–lysosome fusion occurs [150]
dsbAA. castellanni½ log reduction68%Defective oxidative protein folding necessary for replication [151]
icmQU937Incapable of replication100%Defective pore formation in the macrophage [152]
icmRU9371.5 log reduction 97%Defective pore formation in the macrophage [152]
icmSU9371.75 log reduction98%Phagosome–lysosome fusion occurs [152]
icmTU937Incapable of replication100%Phagosome–lysosome fusion occurs [153]
icmWU9372 log reduction99%Phagosome–lysosome fusion occurs [153]
katAA. castellanni2 log reduction99%Susceptible to exogenous hydrogen peroxide [113]
katBA. castellanni2 log reduction99%Susceptible to exogenous hydrogen peroxide [113]
lvgAU93710-fold decrease90%Phagosome–lysosome fusion occurs [150]
lvgAA. castellanni10-fold decrease90%Phagosome–lysosome fusion occurs [150]
1 Expressed in units reported in the original literature. 2 Degradation of Growth Rate refers to the percentage of bacteria decrease within the host cell as compared to the wild-type.
Table 4. Effect of genetic knockdowns on the egress of L. pneumophila within a host cell.
Table 4. Effect of genetic knockdowns on the egress of L. pneumophila within a host cell.
GeneHost CellChange in Egress Rate Efficiency 1Degradation of Egress Rate 2Process
dotAU93775–85%75–85%Defect in inserting pores in eukaryotic membranes [132]
dotBCDU93780%80%Defect in inserting pores in eukaryotic membranes [132]
icmGCDU93770–75%70–75%Defect in inserting pores in eukaryotic membranes [132]
icmJBU93765–80%65–80%Defect in inserting pores in eukaryotic membranes [132]
icmTU93790%90%Defect in pore-formation to egress from cell [153]
legK2A. castellanni1.5 log reduction96.8%Defect in ER recruitment on the LCV [73]
ribU93770–95%70–95%Defective in necrosis-mediated killing of the host cell [121]
ribWI-2685–98%85–98%Defective in necrosis-mediated killing of the host cell [121]
1 Expressed in units reported in the original literature. 2 Degradation of Egress Rate refers to the percentage of bacteria decrease egressing from the host cell as compared to the wild type.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mraz, A.L.; Weir, M.H. Knowledge to Predict Pathogens: Legionella pneumophila Lifecycle Systematic Review Part II Growth within and Egress from a Host Cell. Microorganisms 2022, 10, 141. https://doi.org/10.3390/microorganisms10010141

AMA Style

Mraz AL, Weir MH. Knowledge to Predict Pathogens: Legionella pneumophila Lifecycle Systematic Review Part II Growth within and Egress from a Host Cell. Microorganisms. 2022; 10(1):141. https://doi.org/10.3390/microorganisms10010141

Chicago/Turabian Style

Mraz, Alexis L., and Mark H. Weir. 2022. "Knowledge to Predict Pathogens: Legionella pneumophila Lifecycle Systematic Review Part II Growth within and Egress from a Host Cell" Microorganisms 10, no. 1: 141. https://doi.org/10.3390/microorganisms10010141

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