Open access peer-reviewed chapter

Biofilm, a Cozy Structure for Legionella pneumophila Growth and Persistence in the Environment

Written By

Arwa Abu Khweek and Amal O. Amer

Submitted: 18 April 2019 Reviewed: 13 August 2019 Published: 11 October 2019

DOI: 10.5772/intechopen.89156

From the Edited Volume

Bacterial Biofilms

Edited by Sadik Dincer, Melis Sümengen Özdenefe and Afet Arkut

Chapter metrics overview

784 Chapter Downloads

View Full Metrics

Abstract

Legionella pneumophila (L. pneumophila) is the causative agent of Legionnaires’ disease. Transmission to humans is mediated via inhalation of contaminated water droplets. L. pneumophila is widely distributed in man-made water systems, multiple species of protozoa, and nematodes. L. pneumophila persist within multi-species biofilms that cover surfaces within water systems. Virulence, spread, and resistance to biocides are associated with survival of L. pneumophila within multi-organismal biofilm. Outbreaks of Legionellosis are correlated with the existence of L. pneumophila in biofilms, even after the intensive chemical and physical treatments. Several factors negatively or positively modulate the persistence of L. pneumophila within the microbial consortium-containing L. pneumophila. Biofilm-forming L. pneumophila continue to be a public health and economic burden and directly influence the medical and industrial sectors. Diagnosis and hospitalization of patients and prevention protocols cost governments billions of dollars. Dissecting the biological and environmental factors that promote the persistence and physiological adaptation in biofilms can be fundamental to eliminating and preventing the transmission of L. pneumophila. Herein, we review different factors that promote persistence of L. pneumophila within the biofilm consortium, survival strategies used by the bacteria within biofilm community, gene regulation, and finally challenges associated with biofilm resistance to biocides and anti-Legionella treatments.

Keywords

  • legionella pneumophila
  • biofilm
  • Legionellosis
  • protozoa
  • Caenorhabditis elegans

1. Introduction

L. pneumophila, the causative agent of Legionellosis, as being pathogenic to humans was following an outbreak of pneumonia at a convention of the American Legion in Philadelphia, USA in July 1976 [1]. This pathogen causes a severe form of pneumonia termed Legionnaires’ disease (LD), and less frequently, Pontiac fever, a self-limited flu-like illness. Approximately 90% of LD cases are caused by L. pneumophila. Transmission of L. pneumophila occur primarily through the spread of contaminated aerosols present in cooling towers, condensers, faucets, showers, and hot tubs [2, 3]. Although stringent water quality examinations, the formation of contaminated aerosols remains to be a major problem associated with disease spread [4].

Multiple mechanisms of persistence are harbored by L. pneumophila in various environmental conditions and in humans. Following invasion of amoeba or human macrophages, L. pneumophila form the Legionella-containing vacuole (LCV), which acquires vesicles from early and late endosomes, mitochondria and the endoplasmic reticulum (ER), thus escaping the microbicidal endocytic pathway. Hijacking the endocytic pathway by LCV is fundamental in initiating and maintaining a niche that secure L. pneumophila replication [5, 6]. Importantly, a battery of effector proteins produced by the Dot/Icm type IV secretion system of L. pneumophila. The Dot/Icm secreted effectors are required for successful intracellular replication of L. pneumophila [713]. Like other intracellular bacteria, L. pneumophila switch between a transmissive (virulent) and replicative (non-virulent) biphasic cycles. This switch is essential to ensure bacterial replication in nutrient starved or rich environments and transmit between different niches [14]. Nutrient rich environment is conducive of the replicative phase, where L. pneumophila express few virulence factors. While nutrient deprived environment is promotive of the transmissive phase, especially when the phagosome is unable to support the replication phase of L. pneumophila. Hallmark features of the transmissive phase include, increased motility, expression of plethora of virulence factors, resistance to stressors and egress from the infected host [14]. In the environment, L. pneumophila survive as free living (planktonic) or form bacterial biofilms with other organisms that adhere to surfaces [1520]. Moreover, L. pneumophila is able to differentiate into inert, cyst-like but extremely infectious mature intracellular form (MIF) [21, 22]. Resilience of L. pneumophila extracellularly and under harsh environmental settings is attributed to its ability to exist in viable non-culturable (VBNC) state [23, 24]. Harboring a VBNC mode hinders the detection of many Legionella species. In nature, colonization and persistence is promoted via biofilm formation [25], and survival within freshwater amoeba and C. elegans [5, 26].

Herein, we review factors that mediate biofilm persistence, strategies utilized by the bacteria to become a member of the biofilm consortium and modes of eradicating L. pneumophila biofilm.

1.1 Constituents of L. pneumophila biofilm

L. pneumophila is found as sessile cells associated with biofilms in freshwater environments, [19, 27, 28]. Biofilms mediate bacterial attachments to surfaces and to other pre-attached bacterial communities. Attachment is attained via forming an extracellular matrix (ECM) that is composed mainly of water, proteins, exopolysaccharides, lipids, DNA and RNA, and inorganic compounds [2932]. Three developmental phases are required for biofilm formation. (I) initial attachments to a surface, (II) maturation and extracellular matrix formation, and (III) detachments and dispersion of the bacteria. Biofilms eventually develop into three-dimensional structures containing water channels, which allow bacteria to obtain nutrients, oxygen and get rid of waste products. The behavior of L. pneumophila has mainly been studied in the context of mono- or mixed species biofilms, due to the complexity of biofilm formed in natural environment [1719, 33, 34]. Interestingly, L. pneumophila exhibit minor representation among other species in freshwater and environmental biofilms, [27, 28], and the existence of L. pneumophila may be influenced by other microorganisms in complex biofilms [35]. Some bacterial species positively or negatively affect the persistence of L. pneumophila biofilm [19]. Intriguingly, Klebsiella pneumoniae (K. pneumoniae), Flavobacterium sp., Empedobacter breve, Pseudomonas putidaand Pseudomonas fluorescens positively associated with the long-term persistence of L. pneumophila in biofilms [18, 19, 36]. Other species within biofilms seem to be the provider of capsular polysaccharides, extracellular matrix that support the adherence [3739], or the contributor of growth factors that stimulate growth of L. pneumophila [19]. Pseudomonas aeruginosa (P. aeruginosa), Aeromonas hydrophila, Burkholderia cepacia, Acidovorax sp., and Sphingomonas sp. [40] are among species that antagonize the persistence of L. pneumophila within the biofilm [19]. Inhibition of L. pneumophila biofilm by P. aeruginosa could be a consequence of the effect of homoserine lactone quorums sensing (QS) molecule [41], or production of bacteriocin [40]. Interestingly, L. pneumophila can coexist in biofilm formed by P. aeruginosa and K. pneumoniae indicating that the inhibition of L. pneumophila biofilm formation by P. aeruginosa can be alleviated by the permissive K. pneumoniae [19]. The authors suggest that the growth provided by K. pneumoniae to promote survival of L. pneumophila can at the same time lessen the inhibitory effect by P. aeruginosa [19]. Therefore, the identity, number and nature of interactions between bacterial species (commensalism or interference) can directly affect growth of L. pneumophila within biofilms.

Biofilm formation of L. pneumophila in the laboratory is achieved by growing the bacteria under stringent conditions in nutrient-rich Buffered Yeast Extract medium (BYE) [18, 34]. Different temperatures correlated with different amount, degree of attachment and rate of biofilm formation. Mushroom like structure containing water channels is the hallmark features of biofilms formed at 25°C. In contrast, at 37°C L. pneumophila biofilm is thicker and deficient of water channels observed at 25°C. However, filamentous appearance with mat-like structure has been observed with L. pneumophila grown at 42°C. Studies in our laboratory showed that in contrast to the dotA mutant that lacks the type IV secretion, WT L. pneumophila form biofilm when grown statically at 37°C for 7 days.

Our knowledge is lacking regarding the factors encoded by L. pneumophila that promote the attachment and persistence within multispecies biofilms created by other bacteria.

1.2 Formation of biofilms as a survival niche in oligotrophic environment

Biofilm is extremely nutritious environment that harbors a mixture of living, dead organisms as well as protozoa and bacteria. To be a productive member of the microbial consortium, L. pneumophila has to compete with other bacteria for nutrients in a multispecies biofilm. Therefore, it is essential for L. pneumophila to strive in an environment adjacent to bacterial neighbors that best sustains their growth and survival [42]. Given the fastidious and auxotrophic nature of L. pneumophila, supplementation of the laboratory media with amino acids and iron is essential for growth [43, 44]. However, the ability of L. pneumophila to survive in oligotrophic environments is puzzling and suggests that the bacteria can live on a diet provided by other members in the biofilm community. To overcome the starvation mode in oligotrophic environment, L. pneumophila incorporate in two- and multispecies biofilms. Instead of attaching as a primary colonizer, L. pneumophila use a strategic mode where they dock to a pre-established biofilm, thus mediating bacterial survival and association in the biofilm community [19, 42].

Obtaining the required carbon, nitrogen, and amino acids for replication of L. pneumophila seems to be primarily reliant on necrotrophic feeding on the products of dead bacteria and tissues within the biofilm [35, 36]. Moreover, heterotrophic bacteria support growth of L. pneumophila on media that does not usually support growth because it is deficient in L-cysteine and ferric pyrophosphate [45]. Consistent with this, L. pneumophila showed satellite colonies around some aquatic bacteria including Flavobacterium breve, Pseudomonas spp., Alcaligenes spp., and Acinetobacter spp. Further, L. pneumophila are able to obtain nutrients directly from algae and to grow on the extracellular products produced by cyanobacteria under laboratory conditions [46]. Further, several algae such as Scenedesmus spp., Chlorella spp., and Gloeocystis spp., supported the growth of L. pneumophila in basal salt media [28].

The second mechanism by which L. pneumophila obtain nutrient in biofilms is through amoeba. Amoeba serve as a secure niche that provides the environmental host for survival and replication of Legionella species in the environment [47, 48], and protect the bacteria from antibacterial agents [49]. Importantly, pathogenesis of L. pneumophila is correlated with persistence and adaptation of L. pneumophila in various amoebal hosts, and the nature of protozoal species can directly affect biofilm colonization with L. pneumophila [50, 51]. Indeed, L. pneumophila can parasitize more than 20 species of amoebae, three species of ciliated protozoa and one species of slime mold [52, 53]. Further, multiplication within amoeba mediated increase production of polysaccharides by L. pneumophila, thus enhancing its capacity to establish biofilm [54]. Further, debris from dead amoeba has been shown to support L. pneumophila growth [55], and the biomass of protozoa is directly correlated with outbreaks of L. pneumophila. Moreover, absence of amoeba did not result to an increase in the number of biofilm-associated L. pneumophila. Instead, L. pneumophila can enter the VBNC state to mediate their survival [28]. It has been suggested that metazoan such as the C. elegans could provide a natural host for L. pneumophila [56, 57]. Moreover, L. pneumophila survive within biofilm containing protozoan and C. elegans [58]. Therefore, harnessing nutrient from mixed species biofilms as well as survival in the amoeba and C. elegans enhances the persistence of L. pneumophila. Therefore, diversity of biofilm-associated organisms would provide a various means of nutrient acquisition in oligotrophic environment for such a fastidious organism.

1.3 Factors influencing biofilm formation by L. pneumophila

1.3.1 Cyclic-di-GMP

Regulation of bacterial pathogenesis and biofilm formation has been associated with the bacterial second messenger Cyclic-dimeric diguanylate (c-di-GMP) [5962]. Biofilm regulation for several bacteria has been shown to be reliant on c-di-GMP [6365]. Two main enzymes have been implicated in regulating the synthesis of the c-di-GMP. (I) A diguanylate cyclases (DGCs) containing GGDEF domain mediates the production of c-di-GMP from two GTPs molecules [66]. (II) A phosphodiesterases (PDEs) proteins containing EAL domain that mediate the degradation of c-di-GMP [66].

The L. pneumophila genome encodes for 22–24 GGDEF/EAL-containing proteins that vary between strains, suggesting that c-di-GMP signaling plays a role in the L. pneumophila life style [6769]. Furthermore, L. pneumophila replication within amoeba and macrophages as well as virulence is influenced by the expression of GGDEF/EAL-containing proteins [68, 69]. Three GGDEF/EAL-containing proteins positively regulate biofilm formation in L. pneumophila Lens, [67]. L. pneumophila lacking these proteins showed reduced biofilm formation, however the level of c-di-GMP was not different when compared to the wild type (WT) bacteria [67]. However, two GGDEF/EAL-containing proteins have been shown to negatively regulate biofilm formation and deletion of these proteins resulted in overproduction of biofilm but surprisingly a decrease in the level of the c-di-GMP [67]. Therefore, GGDEF/EAL-containing proteins utilize different mechanisms to regulate biofilm by L. pneumophila when compared to other bacteria.

The Haem Nitric oxide/Oxygen (H-NOX) binding domains family of hemoprotein sensors have been demonstrated to play a role in regulating biofilm formation and the c-di-GMP activity [70]. Intriguingly, L. pneumophila is the only prokaryote found to encode two H-NOX proteins and show widespread of the H-NOX proteins in their genomes. Hyper-biofilm formation phenotype is attributed to deletion of hnox1 without influencing growth of L. pneumophila in nutrient proficient media (BYE), mouse macrophages or Acanthamoeba castellanii. Importantly, a diguanylate cyclase is adjacent to hnox1 and when overexpressed, L. pneumophila exhibits a hyper-biofilm phenotype. Presence of the H-NOX in the NO-bound state inhibited the diguanylate cyclase activity; suggesting that the diguanylate cyclase activity is regulated by NO [70]. Exposure to NO did not result in dispersing the adherent bacteria, but instead the biofilm intensity was increased. The reduced level of c-di-GMP has been associated with the excessive biofilm formation and the c-di-GMP degrading ability could enhance biofilm formation [67]. In the aquatic environment, exposure to NO occurs when L. pneumophila is in close contact to denitrifying bacteria, or when exposed to NO produced by macrophages or protozoa. Therefore, biofilm formation can be regulated by NO sensing.

1.3.2 Iron

Even though it is essential for L. pneumophila growth and replication [7173], the concentration of iron must be stringently regulated, to overcome the toxic effect associated with production of reactive oxygen species (ROS), when used in excessive amount [74, 75]. Biofilm formation is inhibited when a fivefold increase in the concentration of iron pyrophosphate was used [17]. In addition, iron salt has been shown to disturb biofilm formation by other bacteria such including P. aeruginosa [76]. Recently, the effect of iron pyrophosphate and several iron chelators on the persistence of L. pneumophila in mixed biofilm were tested [77]. Chelating ferrous iron dipyridyl, DIP, enhanced the growth of (WT or mutant in iron uptake), suggesting that DIP positively contributes to the persistence of L. pneumophila [77]. Interestingly, DIP has no effect on the bacterial population in biofilm or survival of free-living amoeba in the biofilm and is independent of iron acquisition systems as mutants in iron uptake were not affected by DIP. These data suggest that contribution of DIP to the persistence of L. pneumophila in biofilm is via protecting L. pneumophila from the adverse effects of iron due to a decrease in ROS production [77].

1.3.3 Genetic control

Even though biofilm formation plays a role in the colonization, survival, dissemination and likely the pathogenesis of L. pneumophila [78], the genetic factors and molecular mechanisms involved in this process need to be elucidated. Genes that belong to the putative twin-arginine translocation pathway, which is required for transport of folded proteins across the cytoplasmic membrane, have been shown to be required for biofilm formation. Biofilm formation is reduced in mutants with insertional inactivation of the tatB and tatC genes [79]. Further, biofilm formation in static microtiter plates is impaired in a strain lacking the flagellar sigma factor FliA (σ 28) [18]. Expression of genes associated with the transmissive phase of L. pneumophila is controlled by FliA [80, 81]. Biofilm-derived L. pneumophila down-regulate FliA expression compared to planktonic bacteria in mouse macrophages infection, [82]. Production of flagella is controlled by L. pneumophila quorum sensing (Lqs) signaling compound LAI-1(3-hydroxypentadecane-4-one) as well as the stationary phase regulatory network, sensing availability of nutrient [83]. However, the flagella are not required for attachment and persistence of L. pneumophila biofilm formed by K. pneumonia [19]. This is consistent with our observation showing the down-regulation of the flagella during biofilm formation in mouse macrophage [82].

Binding to sulfated glycosaminoglycans (CAGs) of the host extracellular matrix is mediated via the Legionella collagen-like (LcI) adhesin. Even though LcI is widely distributed in different L. pneumophila environmental and clinical isolates, it is lacking in poor biofilm producers; indicating the acquisition of this gene by horizontal gene transfer to L. pneumophila [84]. The GC content of lpg2644 is different from the rest of L. pneumophila genome [84], indicating the acquisition of this gene by horizontal gene transfer to L. pneumophila [84]. Further, biofilm formation, cell–cell adhesion and cell-matrix interactions is reduced in strains with mutation in lpg2644 [84]. The L. pneumophila lpg2644 gene is differentially regulated during growth phases and biofilm formation [41]. Regulation of late stages of biofilm formation is mediated by P. aeruginosa quorum sensing (3OC12-HSL). Therefore, regulation of biofilm formation promotes dispersion of bacteria and mediates initiation of another biofilm cycle to another surface [41]. These events are crucial for the proliferation and transmission of L. pneumophila [78].

1.3.4 Quorum sensing

In Gram-negative bacteria, gene expression of several bacterial processes, including virulence, sporulation, bioluminescence, competence and biofilm formation is regulated by quorum sensing (QS) [85, 86]. Quorum sensing bacteria are usually identified in man-made water systems and it is well appreciated that QS signaling regulate environmental biofilm production [87]. The LAI-1 (3-hydroxypentadecane-4-one) QS autoinducer is the only (Legionella quorum sensing) Lqs system identified up to date [8891]. The L. pneumophila LAI-1 is detected by the Lqs system which is composed of the autoinducer synthase LqsA, the homologous sensor kinases LqsS and the response regulator LqsR [8890]. The Lqs system of L. pneumophila is homologous to the cqsAS QS of Vibrio cholera, which regulates cell-density, virulence and biofilm formation [85, 92]. Importantly, the L. pneumophila biofilm formation is inhibited by the P. aeruginosa quorum sensing autoinducer (3-oxo-C12-HSL), which down-regulate the expression of lqsR [41, 93]. Therefore, QS could potentially disperse L. pneumophila biofilm during later stages.

1.4 Modulation of gene expression in biofilms

Differential gene expression between planktonic and biofilm forming L. pneumophila was shown through transcriptomic analysis [17]. The gene expression pattern was compared with the replicative and transmissive phases during growth of L. pneumophila in A. castellanii [94]. Importantly, gene expression profile of sessile bacteria is similar to the replicative phase of L. pneumophila. Furthermore, genes that are involved in repressing the transmissive phase were well expressed in the sessile bacteria [17], suggesting that biofilm is a secure niche for L. pneumophila [17]. The pvcAB gene cluster (which is regulated by iron) is among the genes that were highly expressed in the sessile form [17]. The L. pneumophila pvcA and pvcB genes are homologous exhibit homology to the P. aeruginosa proteins PvcA and PvcB and are required for the production of the iron binding protein (siderophore). The pvcA and pvcB in L. pneumophila encode for a siderophore-like molecule, which promote iron sequestration at a sub toxic level. The second gene cluster, including ahpC2 and ahpD, encodes for alkyl hydroperoxide reductases and play a role in protection against oxidative stress [95, 96] displayed the highest induction in biofilm cells [95]. Iron plays a role in the production of reactive oxygen species and the metabolism of iron and oxidative stress is related. Induction of both pvcAB and ahpC2D genes in sessile cells could be utilized to overcome the toxic environment associated with high iron level concentrations.

Further, examining the expression of the macrophage infectivity potentiator (mip) to transcriptionally active L. pneumophila infected in cell culture was used to evaluate the virulence of biofilm-associated L. pneumophila [16]. Expression of mip is required for growth in protozoa and human macrophages [97]. Further, mip expression is up-regulated during the transmissive stages of L. pneumophila life cycle, but downregulated at early stages of infection [98]. At early stages of biofilm formation, which is similar to the replicative phase, expression of mip was constant. However, at later stages of biofilm formation, which is similar to the replicative phase, mip expression was predominately up-regulated [16]. Upregulation of mip expression could be correlated with the switch to the transmissive phase observed in the planktonic form and suggests that biofilm could protect the replicative form of L. pneumophila.

1.5 Biocides treatments of L. pneumophila biofilm and bacterial resistance

L. pneumophila survive in biofilms covering environmental and artificial water systems such as ventilation and conditioning systems [78]. In addition, biofilm-containing L. pneumophila can become a transient or permanent habitat for other relevant microorganisms. Therefore, biofilm-associated organisms can survive for days, weeks or even months depending on the substratum and the environmental factors that stimulate biofilm formation [99, 100]. To restrict L. pneumophila growth, numerous chemical, physical and thermal disinfection methods have been used against L. pneumophila [101]. However, these treatments generally do not result in total elimination of the bacterium, and after a lag period, recolonization occurs as quickly as the treatments are discontinued [35]. Biofilm-associated L. pneumophila is extremely resistant to disinfectants and biocides [101, 102]. Further, exposure of biofilm-encased bacteria to biocides could lead to entry into a viable non-culturable status [103]. Chlorine and its derivatives are the most common biocides used in disinfection protocols and have been shown to be appropriate in eliminating planktonic L. pneumophila but not biofilms [104]. Resistance of L. pneumophila to disinfection is due not only to its capacity to survive within biofilm, but also the bacteria exhibit the intra-amoebal life-style [105, 106]. Therefore, amoeba- associated L. pneumophila are more resistant to disinfection possibly due to differences in membrane chemistry or life cycle stages of this primitive organism [35, 107]. It has been shown that vesicles containing intracellular L. pneumophila released by amoeba are resistant to biocide treatments [108]. Importantly, these vesicles remained viable for few months [109]. Understanding the molecular mechanisms that governs the intra-amoeba related resistance should pave the way for development of new strategies to eradicate L. pneumophila.

Other methods have been used to limit L. pneumophila such as applying heat which has been shown to be effective in reducing the number of bacteria and protozoan trophozoites, but infective against killing cysts [110, 111]. UV radiation is also effective when the bacteria are in direct contact with the radiation [112]. However, higher UV intensities are required to inactivate the protozoa [113]. Other methods have been proposed to control L. pneumophila growth such as controlling the carbon source within anthropogenic water system [114], or addition of phages to control bacterial or specifically L. pneumophila growth. The phage is capable of degrading polysaccharides and therefore destabilizing the biofilm [115, 116]. Furthermore, nanoparticles have been shown to be effective in reduction of L. pneumophila biofilm volume and showed some efficacy against Staphylococcus aureus and Staphylococcus epidermidis biofilms [117119]. Moreover, several natural compounds (biosurfactants, antimicrobial peptides, protein and essential oil) have been shown to exhibit anti-Legionella properties [120]. Collectively, it is necessary to control L. pneumophila growth and their natural hosts to optimize eradication of the bacteria.

Advertisement

2. Conclusions

Several chemical and physical parameters can influence the behavior of L. pneumophila in biofilms, including the surface, the temperature, carbon and metal concentrations, and the presence of biocides [17, 18, 34, 114, 121128]. Biological factors such as being a member of mixed species biofilm or parasitizing free-living amoeba or nematodes influence biofilm formation by L. pneumophila. Biofilm-associated L. pneumophila is resistant to biocides and Legionellosis outbreaks have been attributed to biofilms. Therefore, it is essential to design new remedies for eradication of L. pneumophila biofilm in different environmental settings. Treatment studies should be performed when the bacterium is in its natural host to determine how the bacterium is protected inside the amoeba and if the passages through the natural hosts modify the resistance. Thus, preventing biofilm formation appears as one strategy to reduce water system contamination.

Advertisement

Acknowledgments

Studies in Dr. Amer’s laboratory are supported by The Ohio State University Center for Clinical and Translational Science Longitudinal Pilot Award (CCTS), R21 AI113477, R01 AI24121 and R01 HL127651. Studies in Dr. Abu Khweek’s laboratory are supported by Birzeit University.

Advertisement

Conflict of interest

The authors of the manuscript declare that the submitted work was carried out in the absence of any personal, professional or financial relationships that could potentially be construed as a conflict of interest.

Advertisement

Author contributions

Arwa Abu Khweek wrote the book chapter, and Amal O. Amer edited the manuscript.

References

  1. 1. Fraser DW et al. Legionnaires’ disease: Description of an epidemic of pneumonia. The New England Journal of Medicine. 1977;297(22):1189-1197
  2. 2. Wagner C et al. Collagen binding protein Mip enables legionella pneumophila to transmigrate through a barrier of NCI-H292 lung epithelial cells and extracellular matrix. Cellular Microbiology. 2007;9(2):450-462
  3. 3. Steinert M, Hentschel U, Hacker J. Legionella pneumophila: an aquatic microbe goes astray. FEMS Microbiology Reviews. 2002;26(2):149-162
  4. 4. Fields BS, Benson RF, Besser RE. Legionella and Legionnaires’ disease: 25 years of investigation. Clinical Microbiology Reviews. 2002;15(3):506-526
  5. 5. Isberg RR, O’Connor TJ, Heidtman M. The legionella pneumophila replication vacuole: Making a cosy niche inside host cells. Nature Reviews. Microbiology. 2009;7(1):13-24
  6. 6. de Felipe KS et al. Evidence for acquisition of legionella type IV secretion substrates via interdomain horizontal gene transfer. Journal of Bacteriology. 2005;187(22):7716-7726
  7. 7. Abu Khweek A et al. The Sphingosine-1-phosphate Lyase (LegS2) contributes to the restriction of legionella pneumophila in murine macrophages. PLoS One. 2016;11(1):e0146410
  8. 8. Khweek AA et al. A bacterial protein promotes the recognition of the legionella pneumophila vacuole by autophagy. European Journal of Immunology. 2013;43(5):1333-1344
  9. 9. Losick VP, Isberg RR. NF-kappaB translocation prevents host cell death after low-dose challenge by legionella pneumophila. The Journal of Experimental Medicine. 2006;203(9):2177-2189
  10. 10. de Felipe KS et al. Legionella eukaryotic-like type IV substrates interfere with organelle trafficking. PLoS Pathogens. 2008;4(8):e1000117
  11. 11. Price CT et al. Host proteasomal degradation generates amino acids essential for intracellular bacterial growth. Science. 2011;334(6062):1553-1557
  12. 12. Belyi Y et al. Legionella pneumophila glucosyltransferase inhibits host elongation factor 1A. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(45):16953-16958
  13. 13. Laguna RK et al. A legionella pneumophila-translocated substrate that is required for growth within macrophages and protection from host cell death. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(49):18745-18750
  14. 14. Newton HJ et al. Molecular pathogenesis of infections caused by legionella pneumophila. Clinical Microbiology Reviews. 2010;23(2):274-298
  15. 15. O’Toole G, Kaplan HB, Kolter R. Biofilm formation as microbial development. Annual Review of Microbiology. 2000;54:49-79
  16. 16. Andreozzi E et al. Role of biofilm in protection of the replicative form of legionella pneumophila. Current Microbiology. 2014;69(6):769-774
  17. 17. Hindre T et al. Transcriptional profiling of legionella pneumophila biofilm cells and the influence of iron on biofilm formation. Microbiology. 2008;154(Pt 1):30-41
  18. 18. Mampel J et al. Planktonic replication is essential for biofilm formation by legionella pneumophila in a complex medium under static and dynamic flow conditions. Applied and Environmental Microbiology. 2006;72(4):2885-2895
  19. 19. Stewart CR, Muthye V, Cianciotto NP. Legionella pneumophila persists within biofilms formed by Klebsiella pneumoniae, Flavobacterium sp., and Pseudomonas fluorescens under dynamic flow conditions. PLoS One. 2012;7(11):e50560
  20. 20. Atlas RM. Legionella: From environmental habitats to disease pathology, detection and control. Environmental Microbiology. 1999;1(4):283-293
  21. 21. Berk SG et al. Packaging of live legionella pneumophila into pellets expelled by Tetrahymena spp. does not require bacterial replication and depends on a dot/Icm-mediated survival mechanism. Applied and Environmental Microbiology. 2008;74(7):2187-2199
  22. 22. Faulkner G, Garduno RA. Ultrastructural analysis of differentiation in legionella pneumophila. Journal of Bacteriology. 2002;184(24):7025-7041
  23. 23. Steinert M et al. Resuscitation of viable but nonculturable legionella pneumophila Philadelphia JR32 by Acanthamoeba castellanii. Applied and Environmental Microbiology. 1997;63(5):2047-2053
  24. 24. Garcia MT et al. Acanthamoeba polyphaga resuscitates viable non-culturable legionella pneumophila after disinfection. Environmental Microbiology. 2007;9(5):1267-1277
  25. 25. Valster RM, Wullings BA, van der Kooij D. Detection of protozoan hosts for legionella pneumophila in engineered water systems by using a biofilm batch test. Applied and Environmental Microbiology. 2010;76(21):7144-7153
  26. 26. Horwitz MA. Formation of a novel phagosome by the Legionnaires’ disease bacterium (legionella pneumophila) in human monocytes. The Journal of Experimental Medicine. 1983;158(4):1319-1331
  27. 27. Declerck P et al. Replication of legionella pneumophila in biofilms of water distribution pipes. Microbiological Research. 2009;164(6):593-603
  28. 28. Declerck P. Biofilms: The environmental playground of legionella pneumophila. Environmental Microbiology. 2010;12(3):557-566
  29. 29. Shirtliff ME, Mader JT, Camper AK. Molecular interactions in biofilms. Chemistry & Biology. 2002;9(8):859-871
  30. 30. Sutherland IW. The biofilm matrix--an immobilized but dynamic microbial environment. Trends in Microbiology. 2001;9(5):222-227
  31. 31. Costerton JW. Overview of microbial biofilms. Journal of Industrial Microbiology. 1995;15(3):137-140
  32. 32. Costerton JW et al. Bacterial biofilms in nature and disease. Annual Review of Microbiology. 1987;41:435-464
  33. 33. Pecastaings S et al. Sessile legionella pneumophila is able to grow on surfaces and generate structured monospecies biofilms. Biofouling. 2010;26(7):809-819
  34. 34. Piao Z et al. Temperature-regulated formation of mycelial mat-like biofilms by legionella pneumophila. Applied and Environmental Microbiology. 2006;72(2):1613-1622
  35. 35. Taylor M, Ross K, Bentham R. Legionella, protozoa, and biofilms: Interactions within complex microbial systems. Microbial Ecology. 2009;58(3):538-547
  36. 36. Vervaeren H et al. Introduction of a boost of legionella pneumophila into a stagnant-water model by heat treatment. FEMS Microbiology Ecology. 2006;58(3):583-592
  37. 37. Wu MC et al. Isolation of genes involved in biofilm formation of a Klebsiella pneumoniae strain causing pyogenic liver abscess. PLoS One. 2011;6(8):e23500
  38. 38. Basson A, Flemming LA, Chenia HY. Evaluation of adherence, hydrophobicity, aggregation, and biofilm development of Flavobacterium johnsoniae-like isolates. Microbial Ecology. 2008;55(1):1-14
  39. 39. Kives J, Orgaz B, Sanjose C. Polysaccharide differences between planktonic and biofilm-associated EPS from Pseudomonas fluorescens B52. Colloids and Surfaces. B, Biointerfaces. 2006;52(2):123-127
  40. 40. Guerrieri E et al. Effect of bacterial interference on biofilm development by legionella pneumophila. Current Microbiology. 2008;57(6):532-536
  41. 41. Mallegol J et al. Essential roles and regulation of the legionella pneumophila collagen-like adhesin during biofilm formation. PLoS One. 2012;7(9):e46462
  42. 42. Watnick P, Kolter R. Biofilm, city of microbes. Journal of Bacteriology. 2000;182(10):2675-2679
  43. 43. George JR et al. Amino acid requirements of legionella pneumophila. Journal of Clinical Microbiology. 1980;11(3):286-291
  44. 44. Edelstein PH. Comparative study of selective media for isolation of legionella pneumophila from potable water. Journal of Clinical Microbiology. 1982;16(4):697-699
  45. 45. Wadowsky RM, Yee RB. Satellite growth of legionella pneumophila with an environmental isolate of Flavobacterium breve. Applied and Environmental Microbiology. 1983;46(6):1447-1449
  46. 46. Tison DL et al. Growth of legionella pneumophila in association with blue-green algae (cyanobacteria). Applied and Environmental Microbiology. 1980;39(2):456-459
  47. 47. Rowbotham TJ. Preliminary report on the pathogenicity of legionella pneumophila for freshwater and soil amoebae. Journal of Clinical Pathology. 1980;33(12):1179-1183
  48. 48. Newsome AL et al. Isolation of an amoeba naturally harboring a distinctive legionella species. Applied and Environmental Microbiology. 1998;64(5):1688-1693
  49. 49. Loret JF, Greub G. Free-living amoebae: Biological by-passes in water treatment. International Journal of Hygiene and Environmental Health. 2010;213(3):167-175
  50. 50. Murga R et al. Role of biofilms in the survival of legionella pneumophila in a model potable-water system. Microbiology. 2001;147(Pt 11):3121-3126
  51. 51. Rowbotham TJ. Pontiac fever, amoebae, and legionellae. Lancet. 1981;1(8210):40-41
  52. 52. Hagele S et al. Dictyostelium discoideum: A new host model system for intracellular pathogens of the genus legionella. Cellular Microbiology. 2000;2(2):165-171
  53. 53. Kikuhara H et al. Intracellular multiplication of legionella pneumophila in Tetrahymena thermophila. Journal of UOEH. 1994;16(4):263-275
  54. 54. Bigot R et al. Intra-amoeba multiplication induces chemotaxis and biofilm colonization and formation for legionella. PLoS One. 2013;8(10):e77875
  55. 55. Temmerman R et al. Necrotrophic growth of legionella pneumophila. Applied and Environmental Microbiology. 2006;72(6):4323-4328
  56. 56. Brassinga AK et al. Caenorhabditis is a metazoan host for legionella. Cellular Microbiology. 2010;12(3):343-361
  57. 57. Hellinga JR et al. Identification of vacuoles containing extraintestinal differentiated forms of legionella pneumophila in colonized Caenorhabditis elegans soil nematodes. Microbiology. 2015;4(4):660-681
  58. 58. Rasch J et al. Legionella-protozoa-nematode interactions in aquatic biofilms and influence of Mip on Caenorhabditis elegans colonization. International Journal of Medical Microbiology. 2016;306(6):443-451
  59. 59. Tamayo R, Pratt JT, Camilli A. Roles of cyclic diguanylate in the regulation of bacterial pathogenesis. Annual Review of Microbiology. 2007;61:131-148
  60. 60. Romling U, Galperin MY, Gomelsky M. Cyclic di-GMP: The first 25 years of a universal bacterial second messenger. Microbiology and Molecular Biology Reviews. 2013;77(1):1-52
  61. 61. Martinez-Gil M, Ramos C. Role of cyclic di-GMP in the bacterial virulence and evasion of the plant immunity. Current Issues in Molecular Biology. 2017;25:199-222
  62. 62. Abu Khweek A, Fetherston JD, Perry RD. Analysis of HmsH and its role in plague biofilm formation. Microbiology. 2010;156(Pt 5):1424-1438
  63. 63. Conner JG et al. The ins and outs of cyclic di-GMP signaling in vibrio cholerae. Current Opinion in Microbiology. 2017;36:20-29
  64. 64. Valentini M, Filloux A. Biofilms and cyclic di-GMP (c-di-GMP) Signaling: Lessons from Pseudomonas aeruginosa and other bacteria. The Journal of Biological Chemistry. 2016;291(24):12547-12555
  65. 65. Bobrov AG et al. Systematic analysis of cyclic di-GMP signalling enzymes and their role in biofilm formation and virulence in Yersinia pestis. Molecular Microbiology. 2011;79(2):533-551
  66. 66. Simm R et al. GGDEF and EAL domains inversely regulate cyclic di-GMP levels and transition from sessility to motility. Molecular Microbiology. 2004;53(4):1123-1134
  67. 67. Pecastaings S et al. New insights into legionella pneumophila biofilm regulation by c-di-GMP signaling. Biofouling. 2016;32(8):935-948
  68. 68. Levi A et al. Cyclic diguanylate signaling proteins control intracellular growth of legionella pneumophila. MBio. 2011;2(1):e00316-e00310
  69. 69. Allombert J et al. Three antagonistic cyclic di-GMP-catabolizing enzymes promote differential dot/Icm effector delivery and intracellular survival at the early steps of legionella pneumophila infection. Infection and Immunity. 2014;82(3):1222-1233
  70. 70. Carlson HK, Vance RE, Marletta MA. H-NOX regulation of c-di-GMP metabolism and biofilm formation in legionella pneumophila. Molecular Microbiology. 2010;77(4):930-942
  71. 71. Radtke AL, O’Riordan MX. Intracellular innate resistance to bacterial pathogens. Cellular Microbiology. 2006;8(11):1720-1729
  72. 72. Reeves MW et al. Metal requirements of legionella pneumophila. Journal of Clinical Microbiology. 1981;13(4):688-695
  73. 73. Schaible UE, Kaufmann SH. Iron and microbial infection. Nature Reviews. Microbiology. 2004;2(12):946-953
  74. 74. Andrews SC, Robinson AK, Rodriguez-Quinones F. Bacterial iron homeostasis. FEMS Microbiology Reviews. 2003;27(2-3):215-237
  75. 75. Lemire JA, Harrison JJ, Turner RJ. Antimicrobial activity of metals: Mechanisms, molecular targets and applications. Nature Reviews. Microbiology. 2013;11(6):371-384
  76. 76. Musk DJ, Banko DA, Hergenrother PJ. Iron salts perturb biofilm formation and disrupt existing biofilms of pseudomonas aeruginosa. Chemistry & Biology. 2005;12(7):789-796
  77. 77. Portier E et al. Iron availability modulates the persistence of legionella pneumophila in complex biofilms. Microbes and Environments. 2016;31(4):387-394
  78. 78. Lau HY, Ashbolt NJ. The role of biofilms and protozoa in legionella pathogenesis: Implications for drinking water. Journal of Applied Microbiology. 2009;107(2):368-378
  79. 79. De Buck E et al. Legionella pneumophila Philadelphia-1 tatB and tatC affect intracellular replication and biofilm formation. Biochemical and Biophysical Research Communications. 2005;331(4):1413-1420
  80. 80. Heuner K et al. Influence of the alternative sigma(28) factor on virulence and flagellum expression of legionella pneumophila. Infection and Immunity. 2002;70(3):1604-1608
  81. 81. Molofsky AB, Shetron-Rama LM, Swanson MS. Components of the legionella pneumophila flagellar regulon contribute to multiple virulence traits, including lysosome avoidance and macrophage death. Infection and Immunity. 2005;73(9):5720-5734
  82. 82. Abu Khweek A et al. Biofilm-derived legionella pneumophila evades the innate immune response in macrophages. Frontiers in Cellular and Infection Microbiology. 2013;3:18
  83. 83. Schell U, Simon S, Hilbi H. Inflammasome recognition and regulation of the legionella flagellum. Current Topics in Microbiology and Immunology. 2016;397:161-181
  84. 84. Duncan C et al. Lcl of legionella pneumophila is an immunogenic GAG binding adhesin that promotes interactions with lung epithelial cells and plays a crucial role in biofilm formation. Infection and Immunity. 2011;79(6):2168-2181
  85. 85. Zhu J et al. Quorum-sensing regulators control virulence gene expression in vibrio cholerae. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(5):3129-3134
  86. 86. Ng WL, Bassler BL. Bacterial quorum-sensing network architectures. Annual Review of Genetics. 2009;43:197-222
  87. 87. Shrout JD, Nerenberg R. Monitoring bacterial twitter: Does quorum sensing determine the behavior of water and wastewater treatment biofilms? Environmental Science & Technology. 2012;46(4):1995-2005
  88. 88. Tiaden A et al. The legionella pneumophila response regulator LqsR promotes host cell interactions as an element of the virulence regulatory network controlled by RpoS and LetA. Cellular Microbiology. 2007;9(12):2903-2920
  89. 89. Tiaden A et al. Synergistic contribution of the legionella pneumophila lqs genes to pathogen-host interactions. Journal of Bacteriology. 2008;190(22):7532-7547
  90. 90. Spirig T et al. The legionella autoinducer synthase LqsA produces an alpha-hydroxyketone signaling molecule. The Journal of Biological Chemistry. 2008;283(26):18113-18123
  91. 91. Tiaden A et al. The autoinducer synthase LqsA and putative sensor kinase LqsS regulate phagocyte interactions, extracellular filaments and a genomic island of legionella pneumophila. Environmental Microbiology. 2010;12(5):1243-1259
  92. 92. Miller MB et al. Parallel quorum sensing systems converge to regulate virulence in vibrio cholerae. Cell. 2002;110(3):303-314
  93. 93. Kimura S et al. Pseudomonas aeruginosa las quorum sensing autoinducer suppresses growth and biofilm production in legionella species. Microbiology. 2009;155(Pt 6):1934-1939
  94. 94. Bruggemann H et al. Virulence strategies for infecting phagocytes deduced from the in vivo transcriptional program of legionella pneumophila. Cellular Microbiology. 2006;8(8):1228-1240
  95. 95. Rocha ER, Smith CJ. Role of the alkyl hydroperoxide reductase (ahpCF) gene in oxidative stress defense of the obligate anaerobe bacteroides fragilis. Journal of Bacteriology. 1999;181(18):5701-5710
  96. 96. LeBlanc JJ, Davidson RJ, Hoffman PS. Compensatory functions of two alkyl hydroperoxide reductases in the oxidative defense system of legionella pneumophila. Journal of Bacteriology. 2006;188(17):6235-6244
  97. 97. Cianciotto NP, Fields BS. Legionella pneumophila mip gene potentiates intracellular infection of protozoa and human macrophages. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(11):5188-5191
  98. 98. Wieland H et al. Intracellular multiplication of legionella pneumophila depends on host cell amino acid transporter SLC1A5. Molecular Microbiology. 2005;55(5):1528-1537
  99. 99. Blasco MD, Esteve C, Alcaide E. Multiresistant waterborne pathogens isolated from water reservoirs and cooling systems. Journal of Applied Microbiology. 2008;105(2):469-475
  100. 100. Buse HY et al. Microbial diversities (16S and 18S rRNA gene pyrosequencing) and environmental pathogens within drinking water biofilms grown on the common premise plumbing materials unplasticized polyvinylchloride and copper. FEMS Microbiology Ecology. 2014;88(2):280-295
  101. 101. Kim BR et al. Literature review--efficacy of various disinfectants against legionella in water systems. Water Research. 2002;36(18):4433-4444
  102. 102. Borella P et al. Water ecology of legionella and protozoan: Environmental and public health perspectives. Biotechnology Annual Review. 2005;11:355-380
  103. 103. Giao MS et al. Incorporation of natural uncultivable legionella pneumophila into potable water biofilms provides a protective niche against chlorination stress. Biofouling. 2009;25(4):335-341
  104. 104. Cooper IR, Hanlon GW. Resistance of legionella pneumophila serotype 1 biofilms to chlorine-based disinfection. The Journal of Hospital Infection. 2010;74(2):152-159
  105. 105. Hilbi H, Hoffmann C, Harrison CF. Legionella spp. outdoors: Colonization, communication and persistence. Environmental Microbiology Reports. 2011;3(3):286-296
  106. 106. Steinert M et al. Regrowth of legionella pneumophila in a heat-disinfected plumbing system. Zentralbl Bakteriol. 1998;288(3):331-342
  107. 107. Dupuy M et al. Efficiency of water disinfectants against legionella pneumophila and Acanthamoeba. Water Research. 2011;45(3):1087-1094
  108. 108. Berk SG et al. Production of respirable vesicles containing live legionella pneumophila cells by two Acanthamoeba spp. Applied and Environmental Microbiology. 1998;64(1):279-286
  109. 109. Bouyer S et al. Long-term survival of legionella pneumophila associated with Acanthamoeba castellanii vesicles. Environmental Microbiology. 2007;9(5):1341-1344
  110. 110. Storey MV et al. The efficacy of heat and chlorine treatment against thermotolerant Acanthamoebae and legionellae. Scandinavian Journal of Infectious Diseases. 2004;36(9):656-662
  111. 111. Farhat M et al. Effects of disinfection on legionella spp., eukarya, and biofilms in a hot water system. Applied and Environmental Microbiology. 2012;78(19):6850-6858
  112. 112. Schwartz T, Hoffmann S, Obst U. Formation of natural biofilms during chlorine dioxide and u.v. disinfection in a public drinking water distribution system. Journal of Applied Microbiology. 2003;95(3):591-601
  113. 113. Hijnen WA, Beerendonk EF, Medema GJ. Inactivation credit of UV radiation for viruses, bacteria and protozoan (oo)cysts in water: A review. Water Research. 2006;40(1):3-22
  114. 114. Pang CM, Liu WT. Biological filtration limits carbon availability and affects downstream biofilm formation and community structure. Applied and Environmental Microbiology. 2006;72(9):5702-5712
  115. 115. Lammertyn E et al. Evidence for the presence of legionella bacteriophages in environmental water samples. Microbial Ecology. 2008;56(1):191-197
  116. 116. Hughes KA, Sutherland IW, Jones MV. Biofilm susceptibility to bacteriophage attack: The role of phage-borne polysaccharide depolymerase. Microbiology. 1998;144(Pt 11):3039-3047
  117. 117. Raftery TD et al. Discrete nanoparticles induce loss of legionella pneumophila biofilms from surfaces. Nanotoxicology. 2014;8(5):477-484
  118. 118. Subbiahdoss G et al. Magnetic targeting of surface-modified superparamagnetic iron oxide nanoparticles yields antibacterial efficacy against biofilms of gentamicin-resistant staphylococci. Acta Biomaterialia. 2012;8(6):2047-2055
  119. 119. Taylor EN et al. Superparamagnetic iron oxide nanoparticles (SPION) for the treatment of antibiotic-resistant biofilms. Small. 2012;8(19):3016-3027
  120. 120. Berjeaud JM et al. Legionella pneumophila: The paradox of a highly sensitive opportunistic waterborne pathogen able to persist in the environment. Frontiers in Microbiology. 2016;7:486
  121. 121. Wright JB, Ruseska I, Costerton JW. Decreased biocide susceptibility of adherent legionella pneumophila. The Journal of Applied Bacteriology. 1991;71(6):531-538
  122. 122. Bezanson G et al. In situ colonization of polyvinyl chloride, brass, and copper by legionella pneumophila. Canadian Journal of Microbiology. 1992;38(4):328-330
  123. 123. Turetgen I, Cotuk A. Monitoring of biofilm-associated legionella pneumophila on different substrata in model cooling tower system. Environmental Monitoring and Assessment. 2007;125(1-3):271-279
  124. 124. Rogers J et al. Influence of plumbing materials on biofilm formation and growth of legionella pneumophila in potable water systems. Applied and Environmental Microbiology. 1994;60(6):1842-1851
  125. 125. Donlan RM et al. Legionella pneumophila associated with the protozoan Hartmannella vermiformis in a model multi-species biofilm has reduced susceptibility to disinfectants. Biofouling. 2005;21(1):1-7
  126. 126. Liu Z et al. Effect of flow regimes on the presence of legionella within the biofilm of a model plumbing system. Journal of Applied Microbiology. 2006;101(2):437-442
  127. 127. Lehtola MJ et al. Survival of Mycobacterium avium, legionella pneumophila, Escherichia coli, and caliciviruses in drinking water-associated biofilms grown under high-shear turbulent flow. Applied and Environmental Microbiology. 2007;73(9):2854-2859
  128. 128. van der Kooij D, Veenendaal HR, Scheffer WJ. Biofilm formation and multiplication of legionella in a model warm water system with pipes of copper, stainless steel and cross-linked polyethylene. Water Research. 2005;39(13):2789-2798

Written By

Arwa Abu Khweek and Amal O. Amer

Submitted: 18 April 2019 Reviewed: 13 August 2019 Published: 11 October 2019