Skip to main content
Biofilm logoLink to Biofilm
. 2024 Mar 30;7:100196. doi: 10.1016/j.bioflm.2024.100196

Ecology of Legionella pneumophila biofilms: The link between transcriptional activity and the biphasic cycle

Ana Barbosa a,b, Nuno F Azevedo a,b, Darla M Goeres a,b,c, Laura Cerqueira a,b,
PMCID: PMC11004079  PMID: 38601816

Abstract

There has been considerable discussion regarding the environmental life cycle of Legionella pneumophila and its virulence potential in natural and man-made water systems. On the other hand, the bacterium's morphogenetic mechanisms within host cells (amoeba and macrophages) have been well documented and are linked to its ability to transition from a non-virulent, replicative state to an infectious, transmissive state.

Although the morphogenetic mechanisms associated with the formation and detachment of the L. pneumophila biofilm have also been described, the capacity of the bacteria to multiply extracellularly is not generally accepted. However, several studies have shown genetic pathways within the biofilm that resemble intracellular mechanisms. Understanding the functionality of L. pneumophila cells within a biofilm is fundamental for assessing the ecology and evaluating how the biofilm architecture influences L. pneumophila survival and persistence in water systems. This manuscript provides an overview of the biphasic cycle of L. pneumophila and its implications in associated intracellular mechanisms in amoeba. It also examines the molecular pathways and gene regulation involved in L. pneumophila biofilm formation and dissemination. A holistic analysis of the transcriptional activities in L. pneumophila biofilms is provided, combining the information of intracellular mechanisms in a comprehensive outline. Furthermore, this review discusses the techniques that can be used to study the morphogenetic states of the bacteria within biofilms, at the single cell and population levels.

Keywords: Biofilm ecology, Spatial organization, Cell functionality, Legionella pneumophila, Biofilm physiology

1. Introduction

Legionella is an intracellular Gram-negative facultative pathogen found in natural aquatic environments worldwide (e.g., lakes, rivers, creeks, hot springs, and other bodies of water), associated with freshwater protozoa [[1], [2], [3]]. However, the colonization of artificial water systems like cooling towers, spa pools and showers can create environments suitable for the growth and spread of Legionella, in particular Legionella pneumophila, leading to human infections [1,4,5].

In recent years, much has been inferred about the persistence of Legionella in water systems [6,7]. The opportunistic nature of this bacterium allows the adaptation to various ecosystems, making it a subject of extensive study as a pathogen [2]. Legionella was the etiological agent of numerous outbreaks worldwide, resulting in considerable costs related to hospitalizations and industrial losses [8,9]. Of the 72 Legionella species, most human infections are associated with L. pneumophila [[10], [11], [12]]. L. pneumophila was first identified as being pathogenic to humans after an outbreak of acute pneumonia at a convention of the American Legion in Philadelphia, USA in 1976, and has since been recognized as the main etiologic agent of Pontiac fever and Legionnaire's Disease (LD), two forms of respiratory illnesses [[13], [14], [15]].

One of the largest outbreaks reported in Europe occurred in Portugal in 2020 with 88 confirmed cases of Legionnaire's Disease and 13 deaths linked to industrial cooling towers [16]. Indeed, cooling towers have been identified as one of the sources of contamination [17]. The ubiquity of L. pneumophila and its link to human health underpins the importance of understanding what triggers infection and motivates the bacterium's virulent behaviour. While not straightforward, the answer can likely be found by its complex ecology and resilience, as L. pneumophila can metabolically, physiologically and morphologically adapt as free-living cells, embedded in biofilms or inside host cells (protozoa and macrophages). This enables this pathogen to survive in nutrient-poor environments and inside hosts, as well as denote an increasing resistance to antimicrobials and disinfectants [18,19] (Fig. 1).

Fig. 1.

Fig. 1

Legionella pneumophila within intracellular and extracellular environments. (A) Freshwater ecosystems. 1. Invasion of protozoan host by a free living planktonic cell. 2. After internalization, the bacterium evades the phagosome-lysosome fusion and starts the intracellular multiplication within the Legionella-containing vacuole (LCV) (replicative phase). 3. After the nutrient's conditions become limited, the bacteria switch to a transmissive phase. 4. New cells can lyse the vacuolar membrane and are released in the extracellular environment. Free-living transmissive bacteria can start a new cycle (1), associate with biofilms (5) or persist in the extracellular environment as viable but non-culturable (VBNC). (B). In nutrient-rich medium, L. pneumophila presents a biphasic life cycle, that resembles the replicative (exponential phase (EP) and transmissive intracellular forms (stationary phase (SP)). (C) Release of the mature infectious form (MIF) from the protozoan host can cause Legionnaire's disease through inhalation of aerosols in humans. Created with BioRender.com.

All these physiological transitions imply morphogenetic changes in the bacterial cell wall, shape and motility [20]. In these water niches, the molecular pathways involved in the amoeba intracellular mechanisms, within the replication and transmissibility mechanisms (see below the biphasic cycle section) have been well identified and are linked to the pathogenesis of the bacterium and its ability to invade and delve into host cells [[21], [22], [23]]. Although the regulatory pathways and genes associated with biofilm growth are being documented [24,25], little is known about the ecology of L. pneumophila and the cellular functionality within the biofilm matrix under different conditions. This poses the question: why do biofilm cells trigger bacterial pathogenicity? The behaviour of L. pneumophila in water biofilms can be affected by various physicochemical parameters such as surface, temperature, iron concentration, and the presence of biocides. Additionally, biological factors such as the composition of the biofilm population can either enhance or hinder the bacteria's persistence [24].

At the cellular scale, external factors govern the formation of L. pneumophila biofilm at the transcriptional level [24,26]. In fact, there is a resemblance between biofilm metabolic expression and intracellular survival mechanisms (replicative/transmissive states), and some genes/molecular pathways involved might be associated with the morphological states within a biofilm that may be linked to virulence [27]. While few studies have found evidence to suggest that the bacterium can reproduce extracellularly [[28], [29], [30]], it has been shown that L. pneumophila does use eukaryotic cells to reproduce and complete the cell cycle [[31], [32], [33]]. Besides that, in biofilms, the non-growing cells of L. pneumophila, present metabolic activity and express virulence genes [27].

As such, knowing the spatial location of cells and the phenotypic diversity at the single-cell level [34,35], is of great importance, since deconstructing how biofilm functions in a given environment may be key to understanding what strategies can be used to monitor, control and eradicate biofilms in water systems. Fortunately, the knowledge on biofilm dynamics and complexity at the single-cell level has been greatly evolving in the past years accompanying the advances in technologies at the analytical, molecular and imaging level [36]. This allowed for the study of gene expression (e.g. RT-qPCR) [37], and spatial organization (e.g. GFP staining; FISH techniques) [38] within these microbial communities structures under different environmental circumstances.

Herein, we will start explaining the concepts behind the L. pneumophila biphasic cycle, providing a framework for the associated intracellular mechanisms in amoeba. Afterwards, the molecular pathways and gene regulation involved in L. pneumophila biofilm formation and dissemination into new niches will be explored, harmonizing the acquired knowledge of these regulatory pathways with the biphasic life cycle in host cells. These insights may help to explain not only the virulence state of the bacteria detached from a biofilm, but also contribute to the debate of the possible extracellular replication of L. pneumophila. Finally, state-of-the-art detection technologies that may be used to further understand L. pneumophila ecology in water systems biofilms will be described.

2. L. pneumophila biphasic life cycle

L. pneumophila exhibits a biphasic life cycle, which is controlled by starvation and environmental stress that induces the transition from metabolically active bacteria to motile, stress-resistant, virulent bacteria [20,39,40]. The biphasic cycle is well described in the host cells' intracellular pathway and resembles the growth curve in an experimental scenario [39,41,42] (Fig. 1). In brief, when conditions are favourable in terms of availability of nutrients and living space, such as within amoebas or macrophages, L. pneumophila replicates (exponential growth/replicative state) in a non-motile and non-cytotoxic form. However, when nutrients become limiting, L. pneumophila switches to a transmissive phase (post-exponential/stationary growth), resulting in a flagellated, spore-like, and stress-resistant virulent form that can egress the host cells [43], survive planktonically in the environment and re-establish a replicative niche in protozoa and possibly biofilms. Once in the bulk system, L. pneumophila exhibits high infectivity, mobility, and cyst-like morphology, and can assume a mature intracellular form (MIF) [44,45] as well as a viable but non-culturable (VBNC) form in response to harsh environmental conditions [46,47] (Fig. 1, Fig. 2).

Fig. 2.

Fig. 2

- (A)Legionella pneumophila within multispecies biofilms. I.L. pneumophila adheres to a pre-existing biofilm. II. The cells find a favourable environment which allows them to repress some virulence genes. III. When favourable conditions are depleted, L. pneumophila starts expressing virulence genes and detaches from the biofilm. (B) Intracellular regulation during host protozoan infection. 1. The bacteria adhere to the host cell through phagocytosis mechanisms. 2. The T4BSS effector proteins and other factors allow the establishment of the LCV, and the bacteria initiate the multiplication (replicative phase). 3. When nutrients become limiting, the bacteria switch to a transmissive phase. 4. Bacteria are released into the cytosol, as mature infectious form (MIF) or enter the viable-but-not-culturable (VBNC) form. N: nucleus; GC: Golgi complex; ER: endoplasmic reticulum; M: mitochondria; (+) High concentrations; (−) Low concentrations. Created with BioRender.com.

This complex pleomorphic behaviour is regulated by a set of regulatory systems that control gene expression. Table 1 compiles the main regulatory systems described that play an important role in L. pneumophila infection in amoeba and in the biofilm life cycle. Proteomics and transcriptomics studies revealed key metabolic pathways, common to in vivo infection models and in vitro broth cultures, that dictate the phenotypic shift from the replicative to the transmissive phase [48]. This phenotype transition is coordinated by regulatory systems that control gene expression, such as regulatory proteins (CsrA, RpoS, FliA and FleQ), the LetA/LetS (LetA/S) two-component system (Legionella transmission activator and sensor, respectively) the stringent response metabolites (RelA, SpoT and ‘ppGpp’) and noncoding/small RNA (nc/sRNA) [[49], [50], [51], [52], [53]]. During the replicative phase, genes related to metabolism, amino acid degradation, sugar assimilation, cell division and biosynthetic processes are upregulated. In contrast, when bacteria enter a transmissive phase, genes related to virulence, survival, host attachment and ingress are upregulated, including Icm/Dot type IV secretion system (T4SS) (intracellular multiplication/defective for organelle trafficking), motility machinery (flagellar and type IV pilus genes), and cyclic-di-GMP regulatory proteins [42].

Table 1.

- Main molecules that play a crucial role in Legionella pneumophila infection in amoeba and in the biofilm.

Virulencefactors Cellular function Biofilm stage Amoeba impact Biphasic cyclephaseinvolveda Reference
FleQ (sigma factor σ54) The master regulator of the flagellar regulation cascade Formation Adhesion; Egress (?) Transmissive [52]
FliA (Alternative sigma factor σ28) Important regulatory functions in the flagellar biosynthesis pathway Formation Adhesion; Egress (?) Transmissive [54,55]
TatB and TaC (Putative twin-arginine translocation pathway) Transport of folded proteins across the cytoplasmic membrane Formation Intracellular replication Transmissive (?) [56]
Lcl (Legionella collagen-like protein) Protein that binds to sulfated glucosaminoglycans (CAGs) present in the host extracellular matrix Formation Adhesion Transmissive (?) [[57], [58], [59]]
LadC Putative adenylate cyclase is involved in host cell processes Unknown Adhesion Transmissive (?) [60]
Type IV pili Inner membrane-associated protein Formation Adhesion Transmissive [61]
BffA Involved in the regulation of motility, cellular replication, and virulence Formation Unknown Transmissive (?) [62]
Type II secretion Export of various virulence factors involved in bacterial pathogenicity Formation Intracellular replication Nutrient acquisition during replicative phase [61,64]
Dot/Icm Type 4 Secretion System (T4SS) Modulates host processes including phagosome-lysosome binding Unknown LCV development and intracellular replication Replicative [65,66]
EnhC Immune escape and persistent survival Unknown Intracellular replication Transmissive (?) [67]
LbtA Rhizoferrin biosynthetic gene critical for iron acquisition Formation Intracellular infection Replicative [63]
FeoB Involved in ferrous iron uptake Formation Unknown Replicative (?) [63]
PvcAB and AhpC2D Gene's cluster whose expression is regulated by iron Protection against oxidative stress Intracellular replication Replicative (?) [68]
SidE effector protein family (SidE, SdeA, SdeB, SdeC) Modulation of bacterial infection Unknown Recruitment of the ER vesicles via ER fragmentation to the LCV; Golgi fragmentation Transmissive [68,69]
PmrA/PmrB Virulence regulator Unknown Intracellular replication Transmissive (?) [70]
CsrA (Carbon storage regulator A) Post-transcriptional regulator of gene expression Affects the expression of the regulators FleQ, RpoS, LqsR Intracellular replication; pathogenicity Replicative [40,50,71]
LetA/LetS (Legionella transmission activator and sensor) Activate expression of two small regulatory RNAs, RsmT and RsmZ Involved in CsrA expression Lysosome evasion Transmissive [27,72,73]
RsmT/RsmZ (Regulator of Secondary Metabolism) Relieve the repression of the transmission traits through binding CsrA Unknown Intracellular replication Transmissive [74,75]
Lqs gene cluster Regulates the switch from the replicative to the transmissive/virulent phase, pathogen-host cell interactions, cell motility Could play a role in the dispersion during later stages of biofilm and regulate the phenotypic variation Intracellular replication; motility Transmissive (?) [[76], [77], [78]]
RpoS Virulence regulator Regulation of FliA expression Regulate motility, sodium sensitivity, and evasion of the endocytic pathway Transmissive [53,79,80]
Mip Macrophage infectivity potentiator Upregulated at the end of biofilm formation Virulence Transmissive [[81], [82], [83], [84]]
LepA/LepB Non-lytic release Unknown Egress Transmissive (?) [85]
SpoT and RelA Synthesis of the alarmone ppGpp Unknown Infectivity; motility. Replicative Transmissive [86,87]
c-di-GMP Signalling system involve motility, virulence, the cell cycle, differentiation, and other processes Biofilm formation and dispersal Intracellular replication; motility Transmissive (?) [88]
ppGpp Involved in expressing or repressing some regulators, such as FleQ, FliA, RpoS, LqsR, LetA Unknown Intracellular replication Transmissive [89,90]
a

(?) proposed by the authors after the literature review.

In the next section, the molecular mechanisms involved in the regulation of this biphasic life cycle during the intracellular infection cycle in the protozoa will be deeply explored.

3. L. pneumophila as a successful facultative intracellular pathogen

In the environment, the debate on the L. pneumophila survival and morphological states in biofilms is related to the presence of surrounding amoeba host cells, such as Hartmannella vermiformis and Acanthamoeba castellanii, and this relationship plays a fundamental role in the ecology and pathology of L. pneumophila [91]. L. pneumophila can also survive outside amoeba, as planktonic cells or within biofilms [54,92,93]. Nevertheless, it is generally accepted that pre-existing biofilms are being used by the bacteria as sheltering niches [93] but L. pneumophila requires the protozoa to replicate within water systems [31,33] and this can be the major strategy for their survival in these harsh environments. Because of their near-constant exposure to biofilm-grazing protozoan [91], L. pneumophila has been able to adapt and withstand the eukaryotic cells phagocytosis, enabling them to persist and replicate intracellularly [94,95]. When the cytoplasmic environment conditions become less fortunate, the bacteria tend to evade the host cells back into the bulk system [96]. Then, by aerosolization of water particles, the bacteria can be inhaled and trigger pulmonary infections (Fig. 1). The co-evolution with protozoa prompted the adaptation of L. pneumophila to human defense cells (macrophages), where it seems to use similar intracellular pathways to persist [21,97]. The macrophages intracellular mechanisms and lung infection are extensively reviewed by Khweek et al. (2010), Newton et al. (2010), Brown et al. (2017) [21,98,99].

3.1. The intracellular pathway

Several bacterial factors enhance the initial attachment of L. pneumophila cells to the host (Table 1). Notably, the Lcl protein, which is a collagen-like protein that plays a crucial role in protozoa and macrophages adhesion [57], is also reported to be essential in biofilm formation. It facilitates adhesion to abiotic substrates and participates in biofilm cohesion processes of cell-cell/cell-matrix interactions [58,59]. Upon entering the amoeba through phagocytosis mechanisms [[100], [101], [102]], the bacterium avoids lysosome-mediated degradation, loses its flagella, and forms a unique replication-permissive compartment, called the Legionella-containing vacuole (LCV). This compartment is surrounded by fragments of host cellular structures such as endosomes, and the endoplasmic reticulum. The LCV allows the bacterium to avoid the host's bactericidal mechanisms and provides a suitable environment for replication (replicative state on Fig. 2 B.2) [21,47]. LCV formation is controlled by the Icm/Dot type IV secretion system (T4SS) (intracellular multiplication/defective for organelle trafficking). This system translocate around 300 effector proteins into host cells [65,66]. These proteins signal transduction, cytoskeletal dynamics, and membrane trafficking, controlling every step of the infection process [65,103]. The initial differentiation from a transmissive stage to a replicative stage is induced by the presence of nutrients, such as amino acids, inside this compartment [104]. When nutrients become limited, bacteria enter the stationary phase and consequently upregulate virulence genes. This transition is a highly coordinated process that is initiated upon nutrient limitation (transition from replicative to transmissive state on Fig. 2 B.3). Amino acid starvation triggers the synthesis and accumulation of guanosine 3,5-bispyrophosphate (ppGpp) [105]. The ppGpp plays a crucial role in recruiting sigma factors allowing the activation of genes required for adapting to the new conditions and repressing those that are no longer necessary. In short, the enzymes RelA and SpoT synthesize ppGpp, resulting in changes in gene expression that lead to phenotypic modulation [87,89,106]. The activation of the alternative sigma factor RpoS, an important virulence factor, results in downstream effects that activate the LetA/LetS two-component system [72]. This system upregulates two small non-coding RNAs, which relieve the repression of virulence-related genes by repressing the global repressor CsrA [40]. Moreover, the sigma factor FliA, an important regulatory function in the flagellar biosynthetic pathway, positively affects the establishment of infection by facilitating the encounter with the host cell, enhancing the invasion capacity, as well as the intracellular replication within Dictyostelium discoideum [55,107,108]. Interesting suggestions have been made regarding the role of transcriptional regulators in surface adhesion and biofilm formation, although none of the mutants lacking rpoS, letA, or csrA inhibited biofilm formation in L. pneumophila [54]. In fact, only the mutant lacking flia showed inhibition of biofilm formation in L. pneumophila [54]. After replicative growth (post-exponential phase), when the nutrients of the host cells are exhausted, the bacteria undergo from a metabolically active, non-infectious state to a virulent, transmissive form, completing the biphasic cycle, egressing the amoeba cells and further integrate a new round in the environmental life cycle [20,21,109].

Furthermore, intracellularly, L. pneumophila can differentiate into a spore-like, stress-resistant, virulent, and metabolically dormant mature infectious form (MIF) that is released into the cytosol and from the host cell (Fig. 2 B.4) [18,45,110]. Bacterial release can occur via non-lytic and lytic pathways [85]. Chen et al. (2004) showed that the LepA and LepB proteins are involved in the non-lytic release of infected vesicles by protozoa, via an exocytic pathway [85]. However, lysis can be mediated by enzymes with cytolytic or hemolytic activity, such as regiolysin, RtxA, metalloprotease MspA, phospholipase PlaB, or other phospholipases [111]. Moreover, morphological forms can differentiate into the VBNC form and persist in the environment when present in water for prolonged periods [44]. VBNC cells have the potential to resurrect and become culturable under favourable conditions, such as changes in environmental factors or nutrient availability. In addition, VBNC cells can serve as a reservoir for antibiotic resistance genes that can be transferred to other bacteria in the surrounding environment, contributing to the spread of antibiotic resistance in water systems [112].

4. Biofilms: a shelter for L. pneumophila

The microbial communities in water systems, along with protozoa and other microflora [113], are known to offer protection to L. pneumophila [7,19,114] and the bacteria have developed mechanisms to acquire nutrients through the colonization of multispecies biofilms. Instead of attaching directly to the surface as a primary colonizer, it joins pre-existing biofilm [31,117,118]. The crucial role in the ecology of L. pneumophila is due to its interaction with the natural microflora [119], which may persist both in the presence and absence of amoeba. Some studies ascertain that L. pneumophila can't survive without the presence of amoeba [32,120,121]. A study conducted by Declerck et al. (2009) using a rotating annular reactor to simulate the biofilms in water distribution pipes, found that the presence of A. castellanii in the biofilms composed of Aeromonas hydrophila, Escherichia coli, Flavobacterium breve and Pseudomonas aeruginosa led to a significantly higher invasion of L. pneumophila compared to control experiments where A. castellanii was absent [31]. Nevertheless, another study using a rotating disc reactor to grow biofilms on stainless steel coupons, showed that L. pneumophila was able to persist in biofilms composed of P. aeruginosa, Klebsiella pneumoniae, and Flavobacterium sp., even in the absence of the protozoan host H. vermiformis. The ability of L. pneumophila to replicate was determined by quantifying the loss of plasmid through quantification of the GFP signal. Although, the study does not delve into the detailed processes through which L. pneumophila remains in the biofilm matrix without replication when H. vermiformis is absent [32]. Additionally, these studies do not explore the potential effect of environmental factors, such as temperature and nutrient availability, on the survival and persistence of L. pneumophila in biofilms [31,32,122].

From another standpoint, L. pneumophila seems to persist even without the presence of amoeba by using the matrix via acquiring metabolites from other bacteria in the biofilm [19,32,68]. Adding to the discussion, certain bacteria facilitate the enduring persistence and presence of L. pneumophila in biofilms, whereas others impede its colonization. For example, Pseudomonas fluorescens, Pseudomonas putida, K. pneumoniae and Flavobacterium sp., provide a positive effect that is described as an effect of the production of capsular extracellular matrix material by these microorganisms, as well as the availability of nutrients essential for the survival and growth of L. pneumophila [93]. On the other side, the presence of other species, such as P. aeruginosa, Aeromonas hydrophila, Burkholderia cepacia, Acidovorax sp., and Sphingomonas sp. [119], play an antagonist role. Stewart et al. (2012) reported another interesting finding that L. pneumophila can survive in a three-species biofilm formed by P. aeruginosa and K. pneumoniae [93]. A previous study suggests that P. aeruginosa Quorum Sensing (QS) exerts bacteriostatic and virulence factors by suppressing L. pneumophila growth and biofilm formation [123], and it appears that permissive K. pneumoniae can overcome this inhibitory effect. This complex survival strategy is controlled by external signalling that triggers specific molecular responses and a complex genetic network.

It is worth noting that multiple studies examining L. pneumophila biofilms found in the literature were conducted in vitro, some of them using 12-well polystyrene microtiter plates [93,122] and may not accurately reflect real-world aquatic systems. In fact, in some studies, monospecies biofilm of L. pneumophila were formed in vitro, which has not been proved to exist in natural environments [115,116]. A conceptual scheme is proposed here to outline plausible molecular mechanisms that operate during L. pneumophila presence in biofilms, including adhesion to pre-established systems, persistence, and dispersal/detachment (see Fig. 2). This could be used to systematize the existing information and provide a starting point for understanding the mechanisms involved in L. pneumophila heterogeneity in biofilms.

5. Transcriptomic regulation on L. pneumophila biofilms

It is conceivable that there are similarities between the intracellular and extracellular mechanisms of the bacterium, including the switch between the replicative and transmissive state, and its behaviour when adhering to, persisting in, and releasing from the biofilm. However, there is currently insufficient evidence to support the proposition that morphogenetic changes are similar in both ecosystems. The information compiled on the molecular regulation involved in the L. pneumophila biofilms enabled us to establish a framework of the molecular mechanisms involved in the adhesion, maturation, and detachment of L. pneumophila biofilms (highlighted in Fig. 2).

It is worth noting that some structured genes involved in protozoa invasion, such as the Lcl protein, Type IV pili, and the FliA, were found to play an important role in biofilm adhesion [54,57,58,61,124]. In addition, it has been found that the global regulator, CsrA, which is involved in downregulation of virulent factors during intracellular infection (transmissive phase), is upregulated in sessile cells [68]. Another noteworthy discovery is the upregulation of mip gene expression in the final stage of biofilm as well as in protozoa infection (transmissive phase) [84,125]. These findings suggest that the biofilm provides a favourable environment that protects the replicative state of L. pneumophila. However, there are still unanswered questions regarding the regulation of the maturation and dispersion of the L. pneumophila biofilm. It is important to deepen our knowledge on these topics.

The upcoming sections will explore the molecular regulation involved in the adhesion of L. pneumophila to the biofilm, the different genes present in sessile cells, and the impact of the signalling systems on the architecture and dispersion of the biofilm will be explored.

5.1. Molecular mechanisms involved in L. pneumophila adhesion to biofilms

Upon attachment to a surface, bacteria undergo morphogenetic changes that initiate the sessile lifestyle and the development of a biofilm matrix, which is contingent upon microenvironmental parameters such as microflora, surface type, and fluid properties [126]. Biofilm formation is a tightly regulated process governed by multiple transcriptomic networks, that are modulated by a wide range of external factors and trigger intricate signalling pathways [25,127]. An essential adhesin protein is the Lcl protein that binds to sulfated glucosaminoglycans (CAGs) present in the host extracellular matrix [58] and also facilitates the adhesion to protozoa and macrophages [57]. It is now well-described as playing a vital role in biofilm formation by facilitating attachment to abiotic substrates and participating in cell-cell/cell-matrix interactions, which is essential to the three-dimensional structure of the biofilm [58,59]. Mallegol et al. (2012), demonstrated differential regulation of Lcl during growth phases and biofilm formation in a static biofilm assay. The mutant lacking Lcl showed impaired adhesion, biofilm formation and intercellular interactions. More interestingly, the down-regulation of Lcl may facilitate the dispersal of L. pneumophila to initiate biofilm colonization on another surface [58]. The twin-arginine transport (Tat) secretion is important for the transport of completely folded proteins across the cytoplasmic membrane, and the deletion of tatB and tatC genes results in a significant reduction in biofilm formation suggesting that this system may facilitate the secretion of specific proteins involved in the early stages of biofilm [56]. Additionally, the tatB and tatC mutants exhibit impaired intracellular replication and showed a significant impact on intracellular replication in Acanthamoeba castellanii [56]. As mentioned in section 3.1 the flagellar sigma factor FliA gene is necessary for the expression of genes associated with the transmissive phase of L. pneumophila, including flagella, macrophage infection, and lysosome evasion [124]. Indeed, a mutant lacking fliA showed reduced biofilm formation, suggesting a role for FliA in this process [54]. Type IV pili have also been implicated in L. pneumophila biofilm colonization based on their role in adherence to protozoan cells [128]. Lucas et al. (2006) showed that type IV pili and the pre-pilin peptidase facilitated L. pneumophila colonization of biofilms and the absence of type IV pili resulted in lower attachment levels compared to the wild-type strain. However, the presence of amoeba allowed for attachment and retention of the mutant type IV pili at levels similar to the wild-type strain [61].

5.2. Genes involved in L. pneumophila biofilms

Hindré et al. (2008), conducted a pioneering biofilm transcriptome analysis of L. pneumophila, comparing the replicative and transmissive phases during the growth of L. pneumophila in A. castellanii. The study demonstrated that biofilm may serve as an apt habitat for L. pneumophila, by the expression of genes that repress the transmissive phase in sessile cells [68]. This study showed that the gene encoding the global regulator, CsrA, which is involved in the downregulation of flagellar gene expression and RpoS during a replicative phase [40], was found to be induced in the sessile cells. These findings imply that the regulatory networks are altered in the biofilm in comparison to planktonic life. Additionally, four genes are expressed in the sessile form that comprises two distinct clusters. The PvcAB gene cluster contains the pvcA and pvcB genes, homologous to the pvcA and pvcB in P. aeruginosa which are necessary for the synthesis of the siderophore pyoverdine. In L. pneumophila, this cluster is believed to regulate iron metabolism by controlling concentration and facilitating uptake and sequestration below toxic levels [68]. The other group of genes, alkyl hydroperoxide reductases, ahpD and ahpC2, have been shown to play an influential role in the defence against oxidative stress in the cells of the formed biofilm [68]. Upregulation of ahpD and ahpC2 is documented to be a feature reflecting L. pneumophila in its replicative phase [42], and a study performed by Quan et al. (2020), confirms this upregulation after 24 h of the intracellular growth phase of L. pneumophila within Acanthamoeba. Iron is an essential nutrient for L. pneumophila growth and persistence, which tightly controls biofilm formation [117]. Therefore, the metabolism of iron and oxidative stress is related and the induction of these two clusters in sessile cells may help L. pneumophila against oxidative stress resulting from high iron concentrations. Another interesting study conducted by Lopez et al. (2023) investigated the role of the rhizoferrin biosynthetic gene (lbtA) in L. pneumophila biofilms and infection of host cells. The study also explored the possibility of functional redundancy between the rhizoferrin and the ferrous iron transport pathway (FeoB) in iron acquisition. The lbtA feoB mutant of L. pneumophila was highly defective in forming biofilms on plastic surfaces. This suggests that both the rhizoferrin biosynthesis gene (lbtA) and ferrous iron transport gene (feoB) are critical for biofilm formation. However, the lbtA-containing complement of the mutant was able to restore biofilm formation, indicating that the lbtA gene plays a specific role in this phenotype [63]. Besides that, Marin et al. (2022), identified a specific gene, bffA, in L. pneumophila that appears to be involved in the regulation of motility, biofilm formation, cellular replication, and virulence. The knockout strain lacking bffA showed enhanced biofilm formation, reduced motility, enhanced uptake into amoeba, and altered growth kinetics on solid media. This suggests that bffA plays a role in signalling pathways that govern changes in growth rate and motility in response to environmental conditions [62]. Furthermore, Andreozzi et al. (2014) showed that the expression levels of the macrophage infectivity potentiator (mip) were constant during the early stages and upregulated at the final stage of biofilm formation [84]. These results are similar to the behaviour of L. pneumophila in the infection cycle in human protozoa and macrophages. In fact, mip gene expression is downregulated during the early stages of infection (replicative phase), but upregulated in the final stages during host cell escape (transmissive phase) [125].

5.2.1. Signalling systems that regulate L. pneumophila biofilms

A variety of small signalling molecules generally mediates cell-cell communication [78]. Quorum sensing is the production and release of chemical signal molecules (autoinducers) that control bacterial group behaviour [78,129]. Quorum sensing is the regulation of gene expression of several bacterial processes and behaviour in response to changes in population density, including virulence, sporulation, and biofilm formation/detachment [78,130]. In these communication processes, L. pneumophila uses the Lqs (Legionella quorum sensing) system, which comprises the autoinducer synthase LqsA, the sensor histidine kinases LqsS and LqsT and the response regulator LqsR [[131], [132], [133]]. At high cell density, the autoinducers accumulate and trigger a coordinated response by binding specific receptors [130]. This system responds to LAI-1 (3-hydroxypentadecane-4-one), a molecule involved in the regulation of virulence, cellular phase switch between the replicative/and the transmissive phase, and motility, among others [134]. LAI-1 is an α-hydroxyketone (AHK) and has been identified in L. pneumophila as a quorum-sensing molecule. These molecules have been described as being able to indirectly influence L. pneumophila biofilm colonization, production of extracellular filaments and sedimentation through the Lqs system [135]. Moreover, the bacterium's response to environmental changes is linked to the molecule cyclic di-GMP (c-di-GMP), which is a crucial signalling system involved in various bacterial traits, such as cell cycle, motility, virulence, and biofilm formation and dispersal [[136], [137], [138]]. Planktonic and sessile lifestyles are promoted by low and high intracellular c-di-GMP concentrations, respectively. In fact, c-di-GMP is a key regulator in biofilm dispersion, controlling intracellular levels and influencing enzyme production for matrix degradation, facilitating dispersion. Active dispersion involves reducing c-di-GMP, breaking down the biofilm, and releasing cells [138]. In L. pneumophila, the c-di-GMP is linked to the Lqs system through the pleiotropic transcription factor, LvbR (Legionella virulence and biofilm regulator). This pleiotropic transcription factor is negatively regulated by the sensor kinase LqsS, directly controlling the production of proteins involved in c-di-GMP metabolism, as well as the biofilm architecture and pathogen-host cell interactions [139,140]. Hochstrasser et al. (2022) explored the Lqs-LvbR and c-di-GMP networks in the migration of Acanthamoeba castellanii through Legionella biofilms. The authors conclude that this regulatory network is directly involved in shaping the biofilm architecture, contributing to the formation of either 'patchy' or 'mat-like' structures. The lvbR-deficient strain of L. pneumophila, which has impaired c-di-GMP regulation, accumulates less sessile biomass, and forms homogeneous mat-like structures, leading to altered biofilm architecture [88].

Personnic et al. (2021) showed that the sessile L. pneumophila exhibits phenotypic heterogeneity and was able to form growing and non-growing (“dormant") bacterial populations, controlled by the Lqs system, the transcription factor LvbR and temperature. Interestingly the non-growing sessile cells showed high metabolic activity, expressed virulence genes, and showed tolerance toward antibiotics. These sessile non-growers may lead to a dormant phase, promoting additional long-term survival capacity in the environmental niche and infecting surrounding protozoa [77]. Another interesting study showed that the LvbR acts as a key regulator of biofilm architecture in L. pneumophila, influencing the accumulation of sessile biomass and the formation of compact bacterial aggregates within the biofilm. In fact, the lvbR-deficient strain of L. pneumophila accumulates less sessile biomass and forms homogeneous mat-like structures, while the wild-type strain develops more compact bacterial aggregates [76].

6. Technologies for biofilms transcriptomic analyses

Knowledge of the ecology of bacteria and their three-dimensional positioning within a biofilm has changed considerably over the last few decades, in line with technological developments and the emergence of new tools applied to the science of biofilms, both microscopic imaging and molecular tools applied to systems biology. It is now possible to model the entire three-dimensional biofilm structure, and predict the changes that may occur in response to environmental variations [141]. Nonetheless, it is also possible to perform genotypic and phenotypic analyses of biofilm cells, using “omics” (metagenomics, transcriptomics, and metabolomics) [35,141]. As explored by Azeredo, a deep understanding of the structure of the biofilm as a whole, but also at the single-cell and single-molecule level and of its relationship with the surrounding environment, will open up the development of models that allow reproducible studies, but also its control or eradication [35]. For L. pneumophila the application of these technologies is not prominent, but on the rise. Table 2 outlines the advantages and disadvantages of a few molecular technologies used to study biofilms in L. pneumophila.

Table 2.

- Molecular techniques: brief description of the benefits, downsides, and some biofilms studies on L. pneumophila.

Technique Benefits Downsides References onL.pneumophilabiofilms
qPCRFluorescence detection during PCR reaction Quantification of gene expression levels Disrupting the 3D structure is require, for DNA extraction
Real time detection Susceptible to inhibitory constituents
Multiplex experiments Doesn't distinguish between live or dead cellsa [31,196,197]
Complementary use with Microarray/RNA-Seq results Lack of correspondence between GU units and CFU's, probably due to VBNC cells
CLSM Fluorescent protein (FP)Plasmidic insertion of a gene coding for a FP, that is expressed when activated Real time detection Cells physiology may be altered
No need for 3D Structures destruction Expression levels can hinder fluorescence signal
Genetically encoded Genetic manipulation of bacteria may affect cellular physiology and survival [32,58,63,116,176,198]
2–4 range of colours on multiplex studies Biofilm growth in real environments cannot be monitored
in vitro biofilm studies can be followed
FISHFluorescently labeled oligonucleotide probes that specifically targets RNA molecules Biofilm growth in real environments can be monitored, at well-defined time points Requires sample fixation to enhance probe accessibility which hinders real-time monitoringb
Multiplex studies that can go to dozens of targets discriminated, with the use of spectral imaging (CLASI-FISH and seq-FISH) Probes need to diffuse through biofilm matrixc [31,193,[199], [200], [201]]
Bacteria are not genetically modified Fluorescence signal dependent on the number of ribosomes (rRNA), or expression levels of target genes (mRNA)
Can detect not only rRNA (population level) but also mRNA sequences (single cell level)
a

PMA-based approaches can allow the discrimination between live and dead cells [159].

b

The use of probe delivery techniques, can overcome the need for fixation steps [195].

c

Due to their neutrally charged and synthetic nature, the use of DNA mimics, namely peptide nucleic acid (PNA) may enhance probes diffusion, by the increased resistance to proteases and nucleases and more affinity to the target sequence [178].

Quantitative polymerase chain reaction (qPCR) is a widely used technique for studying and quantifying gene expression. It enables real-time monitoring and fast, high-throughput detection and quantification of target DNA sequences in various matrices [142]. qPCR can also supplement other technologies such as microarrays and RNA-seq analysis, making an important contribution to the study of biofilm ecology [38,68,143,144]. This highlights the importance of using complementary techniques to significantly improve the understanding of biofilms functionality. Furthermore, multiplex optimized qPCR assays are viable for detecting various targets in a sample [145,146]. While this technique is still practiced for analyzing waterborne biofilms at the single-molecule and single-cell level [[147], [148], [149], [150], [151], [152]], as Nisar et al. (2022), that study the association of L. pneumophila with free-living amoeba in domestic and hospital water systems [152] interpreting the results remains challenging, primarily caused by substances present in water which act as qPCR inhibitors, such as debris, metal ions and humic acids [153,154]. In addition, the number of cells may be overestimated as there is no distinction between DNA/RNA from live and dead cells [155,156]. The implementation of propidium monoazide (PMA), which solely penetrates non-viable cells by damaging their membrane and impeding DNA amplification through DNA cross-linking, has advanced the application of this technique [37,[157], [158], [159], [160]]. While substantial research has been made focusing species detection, the research of mRNA transcripts has also been updated, and there are some attempts to standardize extraction, amplification, and quantification protocols [[161], [162], [163], [164]]. Various extraction kits are available, but to ensure optimal bacterial cell lysis and sample purification, an appropriate optimization of the extraction protocols is necessary, especially due to possible interferences from the biofilm that may still be present [164,165]. Nevertheless, the RNA extraction in biofilms may be the limiting factor when evaluating spatial organization due to the necessity to disrupt the biofilm structure. The extraction and amplification of genomic material fails to provide accurate insights into the morphological condition of the bacteria and the underlying three-dimensional microbial structures [34,166].

Allied with the technical progress of 3D imaging techniques, which are predominantly based on high resolution microscopy, such as confocal laser scanning microscopy (CLSM), fluorescence-based methods are employed to decode bacterial species patterns within biofilms at a single-cell and molecular levels [166]. These technologies include the expression of fluorescent proteins inserted into bacteria plasmids and fluorescence in situ hybridization (FISH) [34]. While both technologies are not superiorly used for L. pneumophila, they have already demonstrated their versatility and robustness in biofilm studies (Fig. 3).

Fig. 3.

Fig. 3

Multispecies biofilm visualization using (A) Fluorescent protein labeling (FP) or (B) Fluorescence in situ hybridization (FISH). Brief description of the protocol for each technique. Created with BioRender.com.

The fluorescent proteins (FP) technique is based on the plasmid insertion of a gene encoding an FP, that will be express upon activation [167]. The abovementioned study performed by Murga et al. (2001), applied fluorescent proteins to study the replication capacity of L. pneumophila [32]. The use of various FPs, such as GFP or mCherry enables multiplex studies [168,169]. Additionally, this technology, recognized for its high specificity and ability to explore temporal and spatial changes, has been frequently utilized over the past two decades to research gene expression, strain identification, and measure specific biofilm functions in real-time.

The effectiveness of FPs fluorescence may be hindered by its reliance on a minimum O2 level during the final stage of chromophore maturation. The distribution of O2 in biofilms is uneven since it depends on the balance between gas diffusion through the biofilm and its consumption by the bacteria. This balance determines the spatio-temporal O2 distribution, which varies significantly according to the environmental conditions and biofilm stage. To address the issue, a novel O2-independent small protein tag called FAST (Fluorescence-Activating and absorption-Shifting Tag) has been used, enabling the reporting of gene expression in oxygen-depleted conditions [170,171]. This technique can widely be applied in water to detect specific bacteria or investigate the physiology of the microbial community [[172], [173], [174], [175]]. In the case of L. pneumophila, researchers have drawn parallels between growth within eukaryotic cells and biofilm formation [63,176]. One example of this approach's implementation is laid out in a recent study that investigates the distribution of transmissive L. pneumophila on a single-cell level within LCV during the late stages of infection, using FP production [176]. The authors identified a PflaA-positive transmissive L. pneumophila subpopulation, which is controlled by the Lqs system, and becomes visible at the LCV periphery, facilitating the exit and propagation of bacteria outside of the damaged host cell.

FISH is a technique typically based on the complementary targeting of ribosomal RNA (rRNA) by fluorescently labeled oligonucleotide probes [177,178]. For the efficient access of the probe to the target sequence, the cells must be fixed/permeabilized, yet maintaining the cell integrity [178,179]. After the hybridization, the probe will label the target sequence under stringent conditions, exciting the fluorochrome. Generally, it is also performed a washing step, to remove the unbound probe, clearing samples visualization on the microscope [180,181]. Several probes can be used simultaneously, detecting different targets in multiplex approaches [180,182,183]. Nevertheless, these multiplex approaches can be limited to the number of bandpass filters used in imaging equipment's (the same happen to FP technique). However, several FISH variants may be used to increase the number of targets to dozens or hundreds [166]. In a FISH-based technique for combinatorial labeling and spectral imaging (CLASI-FISH), a unique combination of two or more fluorophores, selected to maximize spectral discrimination, are used in a same probe. The targets can be distinguished by the spectral properties of the combined fluorochromes [[184], [185], [186]]. FISH can also be applied to study the spatial localization of cells in mono or multispecies biofilm [180]. Furthermore, it is possible to use FISH-based techniques (e.g. seqFISH, MER-FISH par-seqFISH) to detect single messenger RNA molecules (mRNA), allowing to categorize the metabolic state of the bacteria in different biofilms layers. The spatial transcriptomics methodology is pacing the FISH arena [38,187,188]. A work on spatial transcriptomic analysis on P. aeruginosa planktonic and sessile cells was published by Dar and co-workers in 2021 [38]. They developed a parallel sequential FISH (par-seqFISH) to capture cells gene expression profiles, maintaining biofilm structure integrity to study cellular physical and biogeographic context. For L. pneumophila, gene expression using FISH is a promising area yet to explore, but until the present day, this technique was only applied for Legionella detection in water samples [189,190], industrial settings [191,192] and biofilms (species detection) [31,193].

These two techniques are viable in biofilm studies owing to their high signal intensity, ability to distinguish among diverse targets in one experiment, versatility, and specificity. However, each method has distinctive features that imply they are suitable for specific circumstances. FP requires genetic modification of cells, thereby restricting its use in experimental biofilm studies. In FISH, the addition of probes to the biofilm eliminates the need for any prior genetic manipulation, enabling natural biofilm monitoring. Conversely, FP allows for real-time assessment without any invasive cell treatment. On the other hand, FISH entails fixation and permeabilization treatments at each time point, to facilitate probe diffusion through biofilm structure and increase nucleic acid probe target affinity. However, a possible way to overcome this is to combine the use of probe delivery systems [194,195] which are capable of fusing with bacterial envelopes in the absence of fixation chemicals, and synthetic nucleic acid mimics (NAMs), such as Peptide Nucleic Acid (PNA) probes. The shorter length compared to DNA/RNA probes, neutral charge, and chemical structure of the PNA may improve diffusion through the biofilm matrix and enhance target accessibility [178,180,181]. To note that both methods can be combined with other techniques (e.g. qPCR, specific matrix components staining) to provide complementary insights into biofilm spatial organization and bacterial physiology under varying circumstances.

7. Conclusions

This review highlights how the understanding of the functionality of a biofilm and the physiology of the cells and their spatial organization is extremely important in order to optimize the means of controlling and eradicating the presence of L. pneumophila in biofilms. This is especially significant in the case of this pathogen, as a lack of efficient water system disinfection can lead to severe respiratory infections through bacterial cells aerosolization.

Resembling the biphasic cycle of replicative and transmissive states in amoeba, the bacteria present different morphogenetic states at several phases of biofilm development. These mechanisms are well-documented in the literature, although the possibility of L. pneumophila reproducing extracellularly in biofilms without a host still needs a deeper investigation, considering the ecology of the bacterium and the impact of different environmental conditions that the L. pneumophila is exposed to in real setting conditions. In this sense, this review provides a holistic description of the different pathways to gain a better understanding of the behaviour of the bacterium as it enters, persists in, or disperses from the biofilm. This study will enhance our comprehension of the bacteria ecological role in aquatic systems. Various methods that can be integrated to explore the genetic expression of the bacterium and its spatial transcriptomics are available, such as qPCR, and 3D imaging techniques allied to fluorescence imaging (FP labeling and FISH).

Funding

This work was financially supported by: e. Biofilm – “Creation of a group of Excellence on Engineered Biofilms” with the Grant Agreement number 101087568, financed by the European Commission in the scope of the Horizon Europe Framework Programme; LEPABE, UIDB/00511/2020 (DOI: 10.54499/UIDB/00511/2020) and UIDP/00511/2020 (DOI: 10.54499/UIDP/00511/2020) and ALiCE, LA/P/0045/2020 (DOI: 10.54499/LA/P/0045/2020), funded by national funds through FCT/MCTES (PIDDAC). Ana Barbosa received a PhD fellowship supported by national funds through FCT (grant reference: 2022.11840. BD).

CRediT authorship contribution statement

Ana Barbosa: Writing – original draft, Investigation. Nuno F. Azevedo: Writing – review & editing, Supervision, Funding acquisition. Darla M. Goeres: Writing – review & editing, Supervision, Funding acquisition. Laura Cerqueira: Writing – review & editing, Writing – original draft, Supervision, Resources, Conceptualization.

Declaration of competing interest

The authors have no competing interests to declare.

Data availability

No data was used for the research described in the article.

References

  • 1.Mercante J.W., Winchell J.M. Current and emerging Legionella diagnostics for laboratory and outbreak investigations. Clin Microbiol Rev. 2015;28(1):95–133. doi: 10.1128/CMR.00029-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Boamah D.K., et al. From many hosts, one accidental pathogen: the diverse Protozoan Hosts of Legionella. Front Cell Infect Microbiol. 2017;7:477. doi: 10.3389/fcimb.2017.00477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mondino S., et al. Legionnaires' disease: State of the art Knowledge of pathogenesis Mechanisms of Legionella. Annu Rev Pathol. 2020;15:439–466. doi: 10.1146/annurev-pathmechdis-012419-032742. [DOI] [PubMed] [Google Scholar]
  • 4.Bedard E., et al. Local adaptation of Legionella pneumophila within a hospital hot water system increases tolerance to copper. Appl Environ Microbiol. 2021;87(10) doi: 10.1128/AEM.00242-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.van Heijnsbergen E., et al. Confirmed and potential sources of Legionella reviewed. Environ Sci Technol. 2015;49(8):4797–4815. doi: 10.1021/acs.est.5b00142. [DOI] [PubMed] [Google Scholar]
  • 6.Falkinham J.O., 3rd, et al. Epidemiology and ecology of opportunistic premise plumbing pathogens: Legionella pneumophila, Mycobacterium avium, and Pseudomonas aeruginosa. Environ Health Perspect. 2015;123(8):749–758. doi: 10.1289/ehp.1408692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhang C., Lu J. Legionella: a promising supplementary Indicator of microbial drinking water Quality in municipal engineered water systems. Front Environ Sci. 2021;9:1–22. doi: 10.3389/fenvs.2021.684319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Collier S.A., et al. Estimate of burden and direct healthcare cost of infectious waterborne disease in the United States. Emerg Infect Dis. 2021;27(1):140–149. doi: 10.3201/eid2701.190676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Prevention, E.C.f.D. and Control . 2019. Legionnaires' disease—annual epidemiological report for. [Google Scholar]
  • 10.Khodr A., et al. Molecular epidemiology, phylogeny and evolution of Legionella. Infect Genet Evol. 2016;43:108–122. doi: 10.1016/j.meegid.2016.04.033. [DOI] [PubMed] [Google Scholar]
  • 11.Muder R.R., Victor L.Y. Infection due to Legionella species other than L. pneumophila. Clin Infect Dis. 2002;35(8):990–998. doi: 10.1086/342884. [DOI] [PubMed] [Google Scholar]
  • 12.Parte A.C., et al. List of prokaryotic names with standing in nomenclature (LPSN) moves to the DSMZ. Int J Syst Evol Microbiol. 2020;70(11):5607–5612. doi: 10.1099/ijsem.0.004332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.McDade J.E., et al. Legionnaires' disease: isolation of a bacterium and demonstration of its role in other respiratory disease. N Engl J Med. 1977;297(22):1197–1203. doi: 10.1056/NEJM197712012972202. [DOI] [PubMed] [Google Scholar]
  • 14.Diederen B. Legionella spp. and Legionnaires' disease. J Infect. 2008;56(1):1–12. doi: 10.1016/j.jinf.2007.09.010. [DOI] [PubMed] [Google Scholar]
  • 15.Glick T.H., et al. Pontiac fever. An epidemic of unknown etiology in a health department: I. Clinical and epidemiologic aspects. Am J Epidemiol. 1978;107(2):149–160. doi: 10.1093/oxfordjournals.aje.a112517. [DOI] [PubMed] [Google Scholar]
  • 16.Almeida D.Q., et al. Outbreak of legionnaires' Disease in the northern Portuguese coast During the COVID-19 pandemic. Acta Med Port. 2021 doi: 10.20344/amp.15823. [DOI] [PubMed] [Google Scholar]
  • 17.Hamilton K.A., et al. Outbreaks of legionnaires' Disease and pontiac fever 2006-2017. Curr Environ Health Rep. 2018;5(2):263–271. doi: 10.1007/s40572-018-0201-4. [DOI] [PubMed] [Google Scholar]
  • 18.Faulkner G., Garduno R.A. Ultrastructural analysis of differentiation in Legionella pneumophila. J Bacteriol. 2002;184(24):7025–7041. doi: 10.1128/JB.184.24.7025-7041.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Berjeaud J.M., et al. Legionella pneumophila: the Paradox of a highly sensitive opportunistic waterborne pathogen Able to Persist in the environment. Front Microbiol. 2016;7:486. doi: 10.3389/fmicb.2016.00486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Oliva G., Sahr T., Buchrieser C. The life cycle of L. pneumophila: cellular differentiation is linked to virulence and metabolism. Front Cell Infect Microbiol. 2018;8:3. doi: 10.3389/fcimb.2018.00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Newton H.J., et al. Molecular pathogenesis of infections caused by Legionella pneumophila. Clin Microbiol Rev. 2010;23(2):274–298. doi: 10.1128/CMR.00052-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yang J.L., Li D., Zhan X.Y. Concept about the virulence factor of Legionella. Microorganisms. 2022;11(1) doi: 10.3390/microorganisms11010074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gomez-Valero L., Buchrieser C. Intracellular parasitism, the driving force of evolution of Legionella pneumophila and the genus Legionella. Gene Immun. 2019;20(5):394–402. doi: 10.1038/s41435-019-0074-z. [DOI] [PubMed] [Google Scholar]
  • 24.Abu Khweek A., Amer A.O. Factors mediating environmental biofilm formation by Legionella pneumophila. Front Cell Infect Microbiol. 2018;8:38. doi: 10.3389/fcimb.2018.00038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Khweek A.A., Amer A.O. Biofilm, a cozy Structure for Legionella pneumophila Growth and Persistence in the environment. in Bacterial Biofilms. 2019 [IntechOpen] [Google Scholar]
  • 26.Toyofuku M., et al. Environmental factors that shape biofilm formation. Biosci Biotechnol Biochem. 2016;80(1):7–12. doi: 10.1080/09168451.2015.1058701. [DOI] [PubMed] [Google Scholar]
  • 27.Chauhan D., Shames S.R. Pathogenicity and Virulence of Legionella: intracellular replication and host response. Virulence. 2021;12(1):1122–1144. doi: 10.1080/21505594.2021.1903199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Surman S., et al. Legionella pneumophila proliferation is not dependent on intracellular replication. Legion. 2001:86–89. [Google Scholar]
  • 29.Surman S., Morton L., Keevil C. The dependence of Legionella pneumophila on other aquatic bacteria for survival on R2A medium. Int Biodeterior Biodegrad. 1994;33(3):223–236. [Google Scholar]
  • 30.Wadowsky R.M., Yee R.B. Satellite growth of Legionella pneumophila with an environmental isolate of Flavobacterium breve. Appl Environ Microbiol. 1983;46(6):1447–1449. doi: 10.1128/aem.46.6.1447-1449.1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Declerck P., et al. Replication of Legionella pneumophila in biofilms of water distribution pipes. Microbiol Res. 2009;164(6):593–603. doi: 10.1016/j.micres.2007.06.001. [DOI] [PubMed] [Google Scholar]
  • 32.Murga R., et al. Role of biofilms in the survival of Legionella pneumophila in a model potable-water system. Microbiology. 2001;147(11):3121–3126. doi: 10.1099/00221287-147-11-3121. [DOI] [PubMed] [Google Scholar]
  • 33.van der Kooij D., et al. Biofilm composition and threshold concentration for growth of Legionella pneumophila on surfaces exposed to flowing warm tap water without disinfectant. Appl Environ Microbiol. 2017;83(5) doi: 10.1128/AEM.02737-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Costa A.M., et al. It is all about location: how to pinpoint microorganisms and their functions in multispecies biofilms. Future Microbiol. 2017;12(11):987–999. doi: 10.2217/fmb-2017-0053. [DOI] [PubMed] [Google Scholar]
  • 35.Azeredo J., et al. Critical review on biofilm methods. Crit Rev Microbiol. 2017;43(3):313–351. doi: 10.1080/1040841X.2016.1208146. [DOI] [PubMed] [Google Scholar]
  • 36.Seneviratne C.J., et al. Multi-omics tools for studying microbial biofilms: current perspectives and future directions. Crit Rev Microbiol. 2020;46(6):759–778. doi: 10.1080/1040841X.2020.1828817. [DOI] [PubMed] [Google Scholar]
  • 37.Magalhães A.P., et al. RNA-based qPCR as a tool to quantify and to characterize dual-species biofilms. Sci Rep. 2019;9(1):13639. doi: 10.1038/s41598-019-50094-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dar D., et al. Spatial transcriptomics of planktonic and sessile bacterial populations at single-cell resolution. Science. 2021;373(6556) doi: 10.1126/science.abi4882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Molofsky A.B., Swanson M.S. Differentiate to thrive: lessons from the Legionella pneumophila life cycle. Mol Microbiol. 2004;53(1):29–40. doi: 10.1111/j.1365-2958.2004.04129.x. [DOI] [PubMed] [Google Scholar]
  • 40.Molofsky A.B., Swanson M.S. Legionella pneumophila CsrA is a pivotal repressor of transmission traits and activator of replication. Mol Microbiol. 2003;50(2):445–461. doi: 10.1046/j.1365-2958.2003.03706.x. [DOI] [PubMed] [Google Scholar]
  • 41.Faucher S.P., Mueller C.A., Shuman H.A. Legionella pneumophila transcriptome during intracellular multiplication in human macrophages. Front Microbiol. 2011;2:60. doi: 10.3389/fmicb.2011.00060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bruggemann H., et al. Virulence strategies for infecting phagocytes deduced from the in vivo transcriptional program of Legionella pneumophila. Cell Microbiol. 2006;8(8):1228–1240. doi: 10.1111/j.1462-5822.2006.00703.x. [DOI] [PubMed] [Google Scholar]
  • 43.Nora T., et al. Molecular mimicry: an important virulence strategy employed by Legionella pneumophila to subvert host functions. Future Microbiol. 2009;4(6):691–701. doi: 10.2217/fmb.09.47. [DOI] [PubMed] [Google Scholar]
  • 44.Robertson P., Abdelhady H., Garduno R.A. The many forms of a pleomorphic bacterial pathogen-the developmental network of Legionella pneumophila. Front Microbiol. 2014;5:670. doi: 10.3389/fmicb.2014.00670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Garduno R.A., et al. Intracellular growth of Legionella pneumophila gives rise to a differentiated form dissimilar to stationary-phase forms. Infect Immun. 2002;70(11):6273–6283. doi: 10.1128/IAI.70.11.6273-6283.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hilbi H., Haas A. Secretive bacterial pathogens and the secretory pathway. Traffic. 2012;13(9):1187–1197. doi: 10.1111/j.1600-0854.2012.01344.x. [DOI] [PubMed] [Google Scholar]
  • 47.Isberg R.R., O'connor T.J., Heidtman M. The Legionella pneumophila replication vacuole: making a cosy niche inside host cells. Nat Rev Microbiol. 2009;7(1):13–24. doi: 10.1038/nrmicro1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ge Z., et al. New global insights on the regulation of the biphasic life cycle and virulence via ClpP-dependent proteolysis in Legionella pneumophila. Mol Cell Proteomics. 2022;21(5):100233. doi: 10.1016/j.mcpro.2022.100233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Edwards R.L., et al. The Legionella pneumophila LetA/LetS two-component system exhibits rheostat-like behavior. Infect Immun. 2010;78(6):2571–2583. doi: 10.1128/IAI.01107-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sahr T., et al. The Legionella pneumophila genome evolved to accommodate multiple regulatory mechanisms controlled by the CsrA-system. PLoS Genet. 2017;13(2) doi: 10.1371/journal.pgen.1006629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.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. Cell Microbiol. 2007;9(12):2903–2920. doi: 10.1111/j.1462-5822.2007.01005.x. [DOI] [PubMed] [Google Scholar]
  • 52.Schulz T., et al. FliA expression analysis and influence of the regulatory proteins RpoN, FleQ and FliA on virulence and in vivo fitness in Legionella pneumophila. Arch Microbiol. 2012;194(12):977–989. doi: 10.1007/s00203-012-0833-y. [DOI] [PubMed] [Google Scholar]
  • 53.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(5):2468–2476. doi: 10.1128/IAI.72.5.2468-2476.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mampel J., et al. Planktonic replication is essential for biofilm formation by Legionella pneumophila in a complex medium under static and dynamic flow conditions. Appl Environ Microbiol. 2006;72(4):2885–2895. doi: 10.1128/AEM.72.4.2885-2895.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Heuner K., et al. Influence of the alternative sigma(28) factor on virulence and flagellum expression of Legionella pneumophila. Infect Immun. 2002;70(3):1604–1608. doi: 10.1128/IAI.70.3.1604-1608.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.De Buck E., et al. Legionella pneumophila Philadelphia-1 tatB and tatC affect intracellular replication and biofilm formation. Biochem Biophys Res Commun. 2005;331(4):1413–1420. doi: 10.1016/j.bbrc.2005.04.060. [DOI] [PubMed] [Google Scholar]
  • 57.Vandersmissen L., et al. A Legionella pneumophila collagen-like protein encoded by a gene with a variable number of tandem repeats is involved in the adherence and invasion of host cells. FEMS Microbiol Lett. 2010;306(2):168–176. doi: 10.1111/j.1574-6968.2010.01951.x. [DOI] [PubMed] [Google Scholar]
  • 58.Mallegol J., et al. Essential roles and regulation of the Legionella pneumophila collagen-like adhesin during biofilm formation. PLoS One. 2012;7(9) doi: 10.1371/journal.pone.0046462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.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. Infect Immun. 2011;79(6):2168–2181. doi: 10.1128/IAI.01304-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Newton H.J., et al. Significant role for ladC in initiation of Legionella pneumophila infection. Infect Immun. 2008;76(7):3075–3085. doi: 10.1128/IAI.00209-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lucas C.E., Brown E., Fields B.S. Type IV pili and type II secretion play a limited role in Legionella pneumophila biofilm colonization and retention. Microbiology (Read) 2006;152(Pt 12):3569–3573. doi: 10.1099/mic.0.2006/000497-0. [DOI] [PubMed] [Google Scholar]
  • 62.Marin C., Kumova O.K., Ninio S. Characterization of a novel regulator of biofilm formation in the pathogen Legionella pneumophila. Biomolecules. 2022;12(2) doi: 10.3390/biom12020225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lopez A.E., et al. Legionella pneumophila rhizoferrin promotes bacterial biofilm formation and growth within amoebae and macrophages. Infect Immun. 2023;91(8) doi: 10.1128/iai.00072-23. 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Graham C.I., et al. Molecular regulation of virulence in Legionella pneumophila. Mol Microbiol. 2023 doi: 10.1111/mmi.15172. [DOI] [PubMed] [Google Scholar]
  • 65.Lifshitz Z., et al. Computational modeling and experimental validation of the Legionella and Coxiella virulence-related type-IVB secretion signal. Proc Natl Acad Sci USA. 2013;110(8):E707–E715. doi: 10.1073/pnas.1215278110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Berger K.H., Isberg R.R. Two distinct defects in intracellular growth complemented by a single genetic locus in Legionella pneumophila. Mol Microbiol. 1993;7(1):7–19. doi: 10.1111/j.1365-2958.1993.tb01092.x. [DOI] [PubMed] [Google Scholar]
  • 67.Liu M., Conover G.M., Isberg R.R. Legionella pneumophila EnhC is required for efficient replication in tumour necrosis factor alpha-stimulated macrophages. Cell Microbiol. 2008;10(9):1906–1923. doi: 10.1111/j.1462-5822.2008.01180.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.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. doi: 10.1099/mic.0.2007/008698-0. [DOI] [PubMed] [Google Scholar]
  • 69.Xie Y., et al. Mechanism and modulation of SidE family proteins in the pathogenesis of Legionella pneumophila. Pathogens. 2023;12(4) doi: 10.3390/pathogens12040629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Al-Khodor S., et al. The PmrA/PmrB two-component system of Legionella pneumophila is a global regulator required for intracellular replication within macrophages and protozoa. Infect Immun. 2009;77(1):374–386. doi: 10.1128/IAI.01081-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Sahr T., et al. Deep sequencing defines the transcriptional map of L. pneumophila and identifies growth phase-dependent regulated ncRNAs implicated in virulence. RNA Biol. 2012;9(4):503–519. doi: 10.4161/rna.20270. [DOI] [PubMed] [Google Scholar]
  • 72.Hammer B.K., Tateda E.S., Swanson M.S. A two-component regulator induces the transmission phenotype of stationary-phase Legionella pneumophila. Mol Microbiol. 2002;44(1):107–118. doi: 10.1046/j.1365-2958.2002.02884.x. [DOI] [PubMed] [Google Scholar]
  • 73.Gal-Mor O., Segal G. The Legionella pneumophila GacA homolog (LetA) is involved in the regulation of icm virulence genes and is required for intracellular multiplication in Acanthamoeba castellanii. Microb Pathog. 2003;34(4):187–194. doi: 10.1016/s0882-4010(03)00027-5. [DOI] [PubMed] [Google Scholar]
  • 74.Rasis M., Segal G. The LetA-RsmYZ-CsrA regulatory cascade, together with RpoS and PmrA, post-transcriptionally regulates stationary phase activation of Legionella pneumophila Icm/Dot effectors. Mol Microbiol. 2009;72(4):995–1010. doi: 10.1111/j.1365-2958.2009.06705.x. [DOI] [PubMed] [Google Scholar]
  • 75.Sahr T., et al. Two small ncRNAs jointly govern virulence and transmission in Legionella pneumophila. Mol Microbiol. 2009;72(3):741–762. doi: 10.1111/j.1365-2958.2009.06677.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hochstrasser R., et al. The pleiotropic Legionella transcription factor LvbR links the Lqs and c-di-GMP regulatory networks to control biofilm architecture and virulence. Environ Microbiol. 2019;21(3):1035–1053. doi: 10.1111/1462-2920.14523. [DOI] [PubMed] [Google Scholar]
  • 77.Personnic N., Striednig B., Hilbi H. Quorum sensing controls persistence, resuscitation, and virulence of Legionella subpopulations in biofilms. ISME J. 2021;15(1):196–210. doi: 10.1038/s41396-020-00774-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Personnic N., Striednig B., Hilbi H. Legionella quorum sensing and its role in pathogen-host interactions. Curr Opin Microbiol. 2018;41:29–35. doi: 10.1016/j.mib.2017.11.010. [DOI] [PubMed] [Google Scholar]
  • 79.Bachman M.A., Swanson M.S. RpoS co-operates with other factors to induce Legionella pneumophila virulence in the stationary phase. Mol Microbiol. 2001;40(5):1201–1214. doi: 10.1046/j.1365-2958.2001.02465.x. [DOI] [PubMed] [Google Scholar]
  • 80.Dong T., Schellhorn H.E. Role of RpoS in virulence of pathogens. Infect Immun. 2010;78(3):887–897. doi: 10.1128/IAI.00882-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Cianciotto N.P., et al. A mutation in the mip gene results in an attenuation of Legionella pneumophila virulence. JID (J Infect Dis) 1990;162(1):121–126. doi: 10.1093/infdis/162.1.121. [DOI] [PubMed] [Google Scholar]
  • 82.Cianciotto N.P., Stamos J.K., Kamp D.W. Infectivity of Legionella pneumophila mip mutant for alveolar epithelial cells. Curr Microbiol. 1995;30:247–250. doi: 10.1007/BF00293641. [DOI] [PubMed] [Google Scholar]
  • 83.Helbig J.H., et al. The PPIase active site of Legionella pneumophila Mip protein is involved in the infection of eukaryotic host cells. 2003 doi: 10.1515/BC.2003.013. [DOI] [PubMed] [Google Scholar]
  • 84.Andreozzi E., et al. Role of biofilm in protection of the replicative form of Legionella pneumophila. Curr Microbiol. 2014;69(6):769–774. doi: 10.1007/s00284-014-0648-y. [DOI] [PubMed] [Google Scholar]
  • 85.Chen J., et al. Legionella effectors that promote nonlytic release from protozoa. Science. 2004;303(5662):1358–1361. doi: 10.1126/science.1094226. [DOI] [PubMed] [Google Scholar]
  • 86.Dalebroux Z.D., Edwards R.L., Swanson M.S. SpoT governs Legionella pneumophila differentiation in host macrophages. Mol Microbiol. 2009;71(3):640–658. doi: 10.1111/j.1365-2958.2008.06555.x. [DOI] [PubMed] [Google Scholar]
  • 87.Zusman T., Gal-Mor O., Segal G. Characterization of a Legionella pneumophila relA insertion mutant and toles of RelA and RpoS in virulence gene expression. J Bacteriol. 2002;184(1):67–75. doi: 10.1128/JB.184.1.67-75.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hochstrasser R., et al. Migration of Acanthamoeba through Legionella biofilms is regulated by the bacterial Lqs-LvbR network, effector proteins and the flagellum. Environ Microbiol. 2022;24(8):3672–3692. doi: 10.1111/1462-2920.16008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Dalebroux Z.D., et al. ppGpp conjures bacterial virulence. Microbiol Mol Biol Rev. 2010;74(2):171–199. doi: 10.1128/MMBR.00046-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Richards A.M., et al. Cellular microbiology and molecular ecology of Legionella-amoeba interaction. Virulence. 2013;4(4):307–314. doi: 10.4161/viru.24290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Shaheen M., Scott C., Ashbolt N.J. Long-term persistence of infectious Legionella with free-living amoebae in drinking water biofilms. Int J Hyg Environ Health. 2019;222(4):678–686. doi: 10.1016/j.ijheh.2019.04.007. [DOI] [PubMed] [Google Scholar]
  • 92.Temmerman R., et al. Necrotrophic growth of Legionella pneumophila. Appl Environ Microbiol. 2006;72(6):4323–4328. doi: 10.1128/AEM.00070-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Stewart C.R., Muthye V., Cianciotto N.P. Legionella pneumophila persists within biofilms formed by Klebsiella pneumoniae, Flavobacterium sp., and Pseudomonas fluorescens under dynamic flow conditions. PLoS One. 2012;7(11) doi: 10.1371/journal.pone.0050560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.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(3):251–265. doi: 10.1046/j.1462-2920.2000.00112.x. [DOI] [PubMed] [Google Scholar]
  • 95.de Felipe K.S., et al. Evidence for acquisition of Legionella type IV secretion substrates via interdomain horizontal gene transfer. J Bacteriol. 2005;187(22):7716–7726. doi: 10.1128/JB.187.22.7716-7726.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Molmeret M., et al. Temporal and spatial trigger of post-exponential virulence-associated regulatory cascades by Legionella pneumophila after bacterial escape into the host cell cytosol. Environ Microbiol. 2010;12(3):704–715. doi: 10.1111/j.1462-2920.2009.02114.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Al-Quadan T., Price C.T., Abu Kwaik Y. Exploitation of evolutionarily conserved amoeba and mammalian processes by Legionella. Trends Microbiol. 2012;20(6):299–306. doi: 10.1016/j.tim.2012.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Khweek A.A., Amer A. Replication of Legionella pneumophila in human cells: why are we susceptible? Front Microbiol. 2010;1:133. doi: 10.3389/fmicb.2010.00133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Brown A.S., et al. The regulation of acute immune responses to the bacterial lung pathogen Legionella pneumophila. J Leukoc Biol. 2017;101(4):875–886. doi: 10.1189/jlb.4MR0816-340R. [DOI] [PubMed] [Google Scholar]
  • 100.King C.H., et al. Effects of cytochalasin D and methylamine on intracellular growth of Legionella pneumophila in amoebae and human monocyte-like cells. Infect Immun. 1991;59(3):758–763. doi: 10.1128/iai.59.3.758-763.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Chang B., et al. Identification of a novel adhesion molecule involved in the virulence of Legionella pneumophila. Infect Immun. 2005;73(7):4272–4280. doi: 10.1128/IAI.73.7.4272-4280.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Prashar A., et al. Mechanism of invasion of lung epithelial cells by filamentousLegionella pneumophila. Cell Microbiol. 2012;14(10):1632–1655. doi: 10.1111/j.1462-5822.2012.01828.x. [DOI] [PubMed] [Google Scholar]
  • 103.Hubber A., Roy C.R. Modulation of host cell function by Legionella pneumophila type IV effectors. Annu Rev Cell Dev Biol. 2010;26:261–283. doi: 10.1146/annurev-cellbio-100109-104034. [DOI] [PubMed] [Google Scholar]
  • 104.Fonseca M.V., Swanson M.S. Nutrient salvaging and metabolism by the intracellular pathogen Legionella pneumophila. Front Cell Infect Microbiol. 2014;4:12. doi: 10.3389/fcimb.2014.00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hammer B.K., Tateda E.S., Swanson M.S. A two-component regulator induces the transmission phenotype of stationary-phase Legionella pneumophila. Mol Microbiol. 2002;44(1):107–118. doi: 10.1046/j.1365-2958.2002.02884.x. [DOI] [PubMed] [Google Scholar]
  • 106.Dalebroux Z.D., et al. Distinct roles of ppGpp and DksA in Legionella pneumophila differentiation. Mol Microbiol. 2010;76(1):200–219. doi: 10.1111/j.1365-2958.2010.07094.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Dietrich C., et al. Flagellum of Legionella pneumophila positively affects the early phase of infection of eukaryotic host cells. Infect Immun. 2001;69(4):2116–2122. doi: 10.1128/IAI.69.4.2116-2122.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Molofsky A.B., Shetron-Rama L.M., Swanson M.S. Components of the Legionella pneumophila flagellar regulon contribute to multiple virulence traits, including lysosome avoidance and macrophage death. Infect Immun. 2005;73(9):5720–5734. doi: 10.1128/IAI.73.9.5720-5734.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.O'Connor T.J., et al. Iron limitation triggers early egress by the intracellular bacterial pathogen Legionella pneumophila. Infect Immun. 2016;84(8):2185–2197. doi: 10.1128/IAI.01306-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Veiga D.F., et al. Predicting transcriptional regulatory interactions with artificial neural networks applied to E. coli multidrug resistance efflux pumps. BMC Microbiol. 2008;8:101. doi: 10.1186/1471-2180-8-101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Eisenreich W., Heuner K. The life stage-specific pathometabolism of Legionella pneumophila. FEBS Lett. 2016;590(21):3868–3886. doi: 10.1002/1873-3468.12326. [DOI] [PubMed] [Google Scholar]
  • 112.Zhang J., et al. Impact of biofilm formation and detachment on the transmission of bacterial antibiotic resistance in drinking water distribution systems. Chemosphere. 2018;203:368–380. doi: 10.1016/j.chemosphere.2018.03.143. [DOI] [PubMed] [Google Scholar]
  • 113.Muhammad M.H., et al. Beyond risk: bacterial biofilms and their regulating approaches. Front Microbiol. 2020;11:928. doi: 10.3389/fmicb.2020.00928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lau H.Y., Ashbolt N.J. The role of biofilms and protozoa in Legionella pathogenesis: implications for drinking water. J Appl Microbiol. 2009;107(2):368–378. doi: 10.1111/j.1365-2672.2009.04208.x. [DOI] [PubMed] [Google Scholar]
  • 115.Pecastaings S., et al. Sessile Legionella pneumophila is able to grow on surfaces and generate structured monospecies biofilms. Biofouling. 2010;26(7):809–819. doi: 10.1080/08927014.2010.520159. [DOI] [PubMed] [Google Scholar]
  • 116.Piao Z., et al. Temperature-regulated formation of mycelial mat-like biofilms by Legionella pneumophila. Appl Environ Microbiol. 2006;72(2):1613–1622. doi: 10.1128/AEM.72.2.1613-1622.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Portier E., et al. Iron availability modulates the persistence of Legionella pneumophila in complex biofilms. Microb Environ. 2016;31(4):387–394. doi: 10.1264/jsme2.ME16010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Declerck P. Biofilms: the environmental playground of Legionella pneumophila. Environ Microbiol. 2010;12(3):557–566. doi: 10.1111/j.1462-2920.2009.02025.x. [DOI] [PubMed] [Google Scholar]
  • 119.Guerrieri E., et al. Effect of bacterial interference on biofilm development by Legionella pneumophila. Curr Microbiol. 2008;57(6):532–536. doi: 10.1007/s00284-008-9237-2. [DOI] [PubMed] [Google Scholar]
  • 120.Donlan R., 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. doi: 10.1080/08927010500044286. [DOI] [PubMed] [Google Scholar]
  • 121.Lau H., Ashbolt N. The role of biofilms and protozoa in Legionella pathogenesis: implications for drinking water. J Appl Microbiol. 2009;107(2):368–378. doi: 10.1111/j.1365-2672.2009.04208.x. [DOI] [PubMed] [Google Scholar]
  • 122.Bigot R., et al. Intra-amoeba multiplication induces chemotaxis and biofilm colonization and formation for Legionella. PLoS One. 2013;8(10) doi: 10.1371/journal.pone.0077875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kimura S., et al. Pseudomonas aeruginosa Las quorum sensing autoinducer suppresses growth and biofilm production in Legionella species. Microbiology. 2009;155(6):1934–1939. doi: 10.1099/mic.0.026641-0. [DOI] [PubMed] [Google Scholar]
  • 124.Heuner K., Steinert M. The flagellum of Legionella pneumophila and its link to the expression of the virulent phenotype. Int J Med Microbiol. 2003;293(2–3):133–143. doi: 10.1078/1438-4221-00259. [DOI] [PubMed] [Google Scholar]
  • 125.Wieland H., et al. Intracellular multiplication of Legionella pneumophila depends on host cell amino acid transporter SLC1A5. Mol Microbiol. 2005;55(5):1528–1537. doi: 10.1111/j.1365-2958.2005.04490.x. [DOI] [PubMed] [Google Scholar]
  • 126.Sauer K., et al. The biofilm life cycle: expanding the conceptual model of biofilm formation. Nat Rev Microbiol. 2022;20(10):608–620. doi: 10.1038/s41579-022-00767-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Abdel-Nour M., et al. Biofilms: the stronghold of Legionella pneumophila. Int J Mol Sci. 2013;14(11):21660–21675. doi: 10.3390/ijms141121660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Stone B.J., Kwaik Y.A. Expression of multiple pili by Legionella pneumophila: identification and characterization of a type IV pilin gene and its role in adherence to mammalian and protozoan cells. Infect Immun. 1998;66(4):1768–1775. doi: 10.1128/iai.66.4.1768-1775.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Miller M.B., Bassler B.L. Quorum sensing in bacteria. Annu Rev Microbiol. 2001;55(1):165–199. doi: 10.1146/annurev.micro.55.1.165. [DOI] [PubMed] [Google Scholar]
  • 130.Rutherford S.T., Bassler B.L. Bacterial quorum sensing: its role in virulence and possibilities for its control. Cold Spring Harb Perspect Med. 2012;2(11) doi: 10.1101/cshperspect.a012427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Spirig T., et al. The Legionella autoinducer synthase LqsA produces an α-hydroxyketone signaling molecule. J Biol Chem. 2008;283(26):18113–18123. doi: 10.1074/jbc.M801929200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.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. Environ Microbiol. 2010;12(5):1243–1259. doi: 10.1111/j.1462-2920.2010.02167.x. [DOI] [PubMed] [Google Scholar]
  • 133.Kessler A., et al. The L egionella pneumophila orphan sensor kinase LqsT regulates competence and pathogen–host interactions as a component of the LAI-1 circuit. Environ Microbiol. 2013;15(2):646–662. doi: 10.1111/j.1462-2920.2012.02889.x. [DOI] [PubMed] [Google Scholar]
  • 134.Schell U., et al. The alpha-hydroxyketone LAI-1 regulates motility, Lqs-dependent phosphorylation signalling and gene expression of Legionella pneumophila. Mol Microbiol. 2016;99(4):778–793. doi: 10.1111/mmi.13265. [DOI] [PubMed] [Google Scholar]
  • 135.Tiaden A., Spirig T., Hilbi H. Bacterial gene regulation by α-hydroxyketone signaling. Trends Microbiol. 2010;18(7):288–297. doi: 10.1016/j.tim.2010.03.004. [DOI] [PubMed] [Google Scholar]
  • 136.Valentini M., Filloux A. Multiple roles of c-di-GMP signaling in bacterial pathogenesis. Annu Rev Microbiol. 2019;73:387–406. doi: 10.1146/annurev-micro-020518-115555. [DOI] [PubMed] [Google Scholar]
  • 137.Hengge R. Principles of c-di-GMP signalling in bacteria. Nat Rev Microbiol. 2009;7(4):263–273. doi: 10.1038/nrmicro2109. [DOI] [PubMed] [Google Scholar]
  • 138.Wille J., Coenye T. Biofilm dispersion: The key to biofilm eradication or opening Pandora's box? Biofilm. 2020;2:100027. doi: 10.1016/j.bioflm.2020.100027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hochstrasser R., Hilbi H. Legionella quorum sensing meets cyclic-di-GMP signaling. Curr Opin Microbiol. 2020;55:9–16. doi: 10.1016/j.mib.2020.01.001. [DOI] [PubMed] [Google Scholar]
  • 140.Hochstrasser R., Hilbi H. Migration of Acanthamoeba castellanii through Legionella biofilms. Methods Mol Biol. 2019;1921:79–89. doi: 10.1007/978-1-4939-9048-1_5. [DOI] [PubMed] [Google Scholar]
  • 141.Raes J., Bork P. Molecular eco-systems biology: towards an understanding of community function. Nat Rev Microbiol. 2008;6(9):693–699. doi: 10.1038/nrmicro1935. [DOI] [PubMed] [Google Scholar]
  • 142.Kralik P., Ricchi M. A basic guide to real time PCR in microbial diagnostics: definitions, parameters, and everything. Front Microbiol. 2017;8:108. doi: 10.3389/fmicb.2017.00108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Crépin A., et al. Sensitivity of Legionella pneumophila to phthalates and their substitutes. Sci Rep. 2023;13(1):22145. doi: 10.1038/s41598-023-49426-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Carlson H.K., Vance R.E., Marletta M.A. H-NOX regulation of c-di-GMP metabolism and biofilm formation in Legionella pneumophila. Mol Microbiol. 2010;77(4):930–942. doi: 10.1111/j.1365-2958.2010.07259.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Marín M.J., et al. Validation of a multiplex qPCR assay for detection and quantification of Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis and Tannerella forsythia in subgingival plaque samples. A comparison with anaerobic culture. Arch Oral Biol. 2019;102:199–204. doi: 10.1016/j.archoralbio.2019.04.014. [DOI] [PubMed] [Google Scholar]
  • 146.Lenz A.P., et al. Localized gene expression in Pseudomonas aeruginosa biofilms. Appl Environ Microbiol. 2008;74(14):4463–4471. doi: 10.1128/AEM.00710-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Li L., et al. Transcriptomic changes of Legionella pneumophila in water. BMC Genom. 2015;16(1):637. doi: 10.1186/s12864-015-1869-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Liang J., Faucher S.P. Transcriptomic adaptation of Legionella pneumophila to transient heat shock. Frontiers in Water. 2022:4. [Google Scholar]
  • 149.Qin Z., et al. Role of autolysin-mediated DNA release in biofilm formation of Staphylococcus epidermidis. Microbiology. 2007;153(7):2083–2092. doi: 10.1099/mic.0.2007/006031-0. [DOI] [PubMed] [Google Scholar]
  • 150.Wolf-Baca M., Siedlecka A. Detection of pathogenic bacteria in hot tap water using the qPCR method: preliminary research. SN Appl Sci. 2019;1:1–9. [Google Scholar]
  • 151.Koh W., et al. Multiplication of the waterborne pathogen Cryptosporidium parvum in an aquatic biofilm system. Parasites Vectors. 2013;6:1–11. doi: 10.1186/1756-3305-6-270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Nisar M.A., et al. Molecular screening and characterization of Legionella pneumophila associated free-living amoebae in domestic and hospital water systems. Water Res. 2022;226:119238. doi: 10.1016/j.watres.2022.119238. [DOI] [PubMed] [Google Scholar]
  • 153.Nappier S.P., et al. Advancements in mitigating interference in quantitative polymerase chain reaction (qPCR) for microbial water quality monitoring. Sci Total Environ. 2019;671:732–740. doi: 10.1016/j.scitotenv.2019.03.242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Schrader C., et al. PCR inhibitors–occurrence, properties and removal. J Appl Microbiol. 2012;113(5):1014–1026. doi: 10.1111/j.1365-2672.2012.05384.x. [DOI] [PubMed] [Google Scholar]
  • 155.Donohue M.J. Quantification of Legionella pneumophila by qPCR and culture in tap water with different concentrations of residual disinfectants and heterotrophic bacteria. Sci Total Environ. 2021;774:145142. doi: 10.1016/j.scitotenv.2021.145142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Falzone L., et al. Droplet digital PCR for the detection and monitoring of Legionella pneumophila. Int J Mol Med. 2020;46(5):1777–1782. doi: 10.3892/ijmm.2020.4724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Krüger N.-J., et al. “Limits of control”–crucial parameters for a reliable quantification of viable campylobacter by real-time PCR. PLoS One. 2014;9(2) doi: 10.1371/journal.pone.0088108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Yasunaga A., et al. Monitoring the prevalence of viable and dead cariogenic bacteria in oral specimens and in vitro biofilms by qPCR combined with propidium monoazide. BMC Microbiol. 2013;13(1):1–9. doi: 10.1186/1471-2180-13-157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Àlvarez G., et al. Method to quantify live and dead cells in multi-species oral biofilm by real-time PCR with propidium monoazide. Amb Express. 2013;3:1–8. doi: 10.1186/2191-0855-3-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Gensberger E.T., Sessitsch A., Kostić T. Propidium monoazide–quantitative polymerase chain reaction for viable Escherichia coli and Pseudomonas aeruginosa detection from abundant background microflora. Anal Biochem. 2013;441(1):69–72. doi: 10.1016/j.ab.2013.05.033. [DOI] [PubMed] [Google Scholar]
  • 161.França A., Melo L.D., Cerca N. Comparison of RNA extraction methods from biofilm samples of Staphylococcus epidermidis. BMC Res Notes. 2011;4(1):1–5. doi: 10.1186/1756-0500-4-572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.França A., Cerca N. Springer; 2023. Gene expression quantification from pathogenic bacterial biofilms by quantitative PCR, in PCR: Methods and Protocols; pp. 133–149. [DOI] [PubMed] [Google Scholar]
  • 163.Fleige S., et al. Comparison of relative mRNA quantification models and the impact of RNA integrity in quantitative real-time RT-PCR. Biotechnol Lett. 2006;28:1601–1613. doi: 10.1007/s10529-006-9127-2. [DOI] [PubMed] [Google Scholar]
  • 164.França A., Bento J.C., Cerca N. Variability of RNA quality extracted from biofilms of foodborne pathogens using different kits impacts mRNA quantification by qPCR. Curr Microbiol. 2012;65:54–59. doi: 10.1007/s00284-012-0124-5. [DOI] [PubMed] [Google Scholar]
  • 165.Franca A., et al. Optimizing a qPCR gene expression quantification assay for S. epidermidis biofilms: a comparison between commercial kits and a customized protocol. PLoS One. 2012;7(5) doi: 10.1371/journal.pone.0037480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Barbosa A., et al. Imaging biofilms using fluorescence in situ hybridization: seeing is believing. Front Cell Infect Microbiol. 2023;13:1195803. doi: 10.3389/fcimb.2023.1195803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Snapp E. Design and use of fluorescent fusion proteins in cell biology. Curr Protoc Cell Biol. 2005;27(1) doi: 10.1002/0471143030.cb2104s27. 21.4. 1-21.4. 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Hansen M.F., et al. Fluidic resistance control enables high-throughput establishment of mixed-species biofilms. Biotechniques. 2019;66(5):235–239. doi: 10.2144/btn-2018-0150. [DOI] [PubMed] [Google Scholar]
  • 169.Santos S.B., et al. Bacteriophage-receptor binding proteins for multiplex detection of Staphylococcus and Enterococcus in blood. Biotechnol Bioeng. 2020;117(11):3286–3298. doi: 10.1002/bit.27489. [DOI] [PubMed] [Google Scholar]
  • 170.Monmeyran A., et al. Four species of bacteria deterministically assemble to form a stable biofilm in a millifluidic channel. npj Biofilms and Microbiomes. 2021;7(1):64. doi: 10.1038/s41522-021-00233-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Monmeyran A., et al. The inducible chemical-genetic fluorescent marker FAST outperforms classical fluorescent proteins in the quantitative reporting of bacterial biofilm dynamics. Sci Rep. 2018;8(1):10336. doi: 10.1038/s41598-018-28643-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Schaefer L.M., Brözel V.S., Venter S.N. Fate of Salmonella Typhimurium in laboratory-scale drinking water biofilms. J Water Health. 2013;11(4):629–635. doi: 10.2166/wh.2013.208. [DOI] [PubMed] [Google Scholar]
  • 173.Waegenaar F., et al. Unwanted coliforms can hide in mature drinking water biofilms, grown in full-scale distribution networks. bioRxiv. 2023:2023. 09. 21.558492. [Google Scholar]
  • 174.Puri D., Fang X., Allison K.R. Fluorescence-based protocol for revealing cellular arrangement in biofilms. STAR protocols. 2023;4(2):102270. doi: 10.1016/j.xpro.2023.102270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Li J., McLellan S., Ogawa S. Accumulation and fate of green fluorescent labeled Escherichia coli in laboratory-scale drinking water biofilters. Water Res. 2006;40(16):3023–3028. doi: 10.1016/j.watres.2006.06.029. [DOI] [PubMed] [Google Scholar]
  • 176.Striednig B., et al. Quorum sensing governs a transmissive Legionella subpopulation at the pathogen vacuole periphery. EMBO Rep. 2021;22(9) doi: 10.15252/embr.202152972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Amann R.I., et al. Combination of 16S rRNA-targeted oligonucleotide probes with flow cytometry for analyzing mixed microbial populations. Appl Environ Microbiol. 1990;56(6):1919–1925. doi: 10.1128/aem.56.6.1919-1925.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Nacher-Vazquez M., et al. The role of Nucleic Acid Mimics (NAMs) on FISH-based techniques and applications for microbial detection. Microbiol Res. 2022;262:127086. doi: 10.1016/j.micres.2022.127086. [DOI] [PubMed] [Google Scholar]
  • 179.Cerqueira L., et al. DNA mimics for the rapid identification of microorganisms by fluorescence in situ hybridization (FISH) Int J Mol Sci. 2008;9(10):1944–1960. doi: 10.3390/ijms9101944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Almeida C., et al. Discriminating multi-species populations in biofilms with peptide nucleic acid fluorescence in situ hybridization (PNA FISH) PLoS One. 2011;6(3) doi: 10.1371/journal.pone.0014786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Cerqueira L., et al. Biofilm formation with mixed cultures of Pseudomonas aeruginosa/Escherichia coli on silicone using artificial urine to mimic urinary catheters. Biofouling. 2013;29(7):829–840. doi: 10.1080/08927014.2013.807913. [DOI] [PubMed] [Google Scholar]
  • 182.Cerqueira L., et al. Validation of a fluorescence in situ hybridization method using peptide nucleic acid probes for detection of Helicobacter pylori clarithromycin resistance in gastric biopsy specimens. J Clin Microbiol. 2013;51(6):1887–1893. doi: 10.1128/JCM.00302-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Thurnheer T., Gmür R., Guggenheim B. Multiplex FISH analysis of a six-species bacterial biofilm. J Microbiol Methods. 2004;56(1):37–47. doi: 10.1016/j.mimet.2003.09.003. [DOI] [PubMed] [Google Scholar]
  • 184.Azevedo A.S., et al. Spectral imaging and nucleic acid mimics fluorescence in situ hybridization (SI-NAM-FISH) for multiplex detection of clinical pathogens. Front Microbiol. 2022;13:976639. doi: 10.3389/fmicb.2022.976639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Valm A.M., Welch J.L.M., Borisy G.G. CLASI-FISH: principles of combinatorial labeling and spectral imaging. Syst Appl Microbiol. 2012;35(8):496–502. doi: 10.1016/j.syapm.2012.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Valm A.M., et al. Systems-level analysis of microbial community organization through combinatorial labeling and spectral imaging. Proc Natl Acad Sci USA. 2011;108(10):4152–4157. doi: 10.1073/pnas.1101134108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Chen K.H., et al. Spatially resolved, highly multiplexed RNA profiling in single cells. Science. 2015;348(6233) doi: 10.1126/science.aaa6090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Lubeck E., et al. Single-cell in situ RNA profiling by sequential hybridization. Nat Methods. 2014;11(4):360–361. doi: 10.1038/nmeth.2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Moreno Y., Moreno-Mesonero L., García-Hernández J. DVC-FISH to identify potentially pathogenic Legionella inside free-living amoebae from water sources. Environ Res. 2019;176:108521. doi: 10.1016/j.envres.2019.06.002. [DOI] [PubMed] [Google Scholar]
  • 190.Nácher-Vázquez M., et al. Development of a novel peptide nucleic acid probe for the detection of Legionella spp. in water samples. Microorganisms. 2022;10(7):1409. doi: 10.3390/microorganisms10071409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Kirschner A.K., et al. Development of a new CARD-FISH protocol for quantification of Legionella pneumophila and its application in two hospital cooling towers. J Appl Microbiol. 2012;112(6):1244–1256. doi: 10.1111/j.1365-2672.2012.05289.x. [DOI] [PubMed] [Google Scholar]
  • 192.Zeybek Z., Gungor N.D., Turetgen I. Investigation of heterotrophic bacteria, legionella and free-living amoeba in cooling tower samples by fish and culture methods. European Journal of Biology. 2017;76(1):7–13. [Google Scholar]
  • 193.Wilks S.A., Keevil C.W. Targeting species-specific low-affinity 16S rRNA binding sites by using peptide nucleic acids for detection of Legionellae in biofilms. Appl Environ Microbiol. 2006;72(8):5453–5462. doi: 10.1128/AEM.02918-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Zhang Y., et al. Multi-targeted antisense oligonucleotide delivery by a framework nucleic acid for inhibiting biofilm formation and virulence. Nano-Micro Lett. 2020;12:1–13. doi: 10.1007/s40820-020-0409-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Moreira L., et al. Liposome delivery of nucleic acids in bacteria: toward in vivo labeling of human microbiota. ACS Infect Dis. 2022;8(7):1218–1230. doi: 10.1021/acsinfecdis.1c00601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Taylor M.J., Bentham R.H., Ross K.E. Limitations of using propidium monoazide with qPCR to discriminate between live and dead Legionella in biofilm samples. Microbiol Insights. 2014;7 doi: 10.4137/MBI.S17723. p. MBI. S17723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Buse H.Y., et al. Effect of temperature and colonization of Legionella pneumophila and Vermamoeba vermiformis on bacterial community composition of copper drinking water biofilms. Microb Biotechnol. 2017;10(4):773–788. doi: 10.1111/1751-7915.12457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Buchrieser C., Hilbi H. Legion: Methods and Protocols. 2019 [Google Scholar]
  • 199.Declerck P., Ollevier F. Detection of Legionella in various sample types using whole-cell fluorescent in situ hybridization. Diagnostic Bacteriology Protocols. 2006:175–183. doi: 10.1385/1-59745-143-6:175. [DOI] [PubMed] [Google Scholar]
  • 200.Declerck P., et al. Replication of Legionella pneumophila in floating biofilms. Curr Microbiol. 2007;55(5):435–440. doi: 10.1007/s00284-007-9006-7. [DOI] [PubMed] [Google Scholar]
  • 201.Gião M.S., et al. Interaction of Legionella pneumophila and Helicobacter pylori with bacterial species isolated from drinking water biofilms. BMC Microbiol. 2011;11:1–10. doi: 10.1186/1471-2180-11-57. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data was used for the research described in the article.


Articles from Biofilm are provided here courtesy of Elsevier

RESOURCES