Abstract

Pseudomonas aeruginosa is a metabolically versatile bacterium that is found in a wide range of biotic and abiotic habitats. It is a major human opportunistic pathogen causing numerous acute and chronic infections. The critical traits contributing to the pathogenic potential of P. aeruginosa are the production of a myriad of virulence factors, formation of biofilms and antibiotic resistance. Expression of these traits is under stringent regulation, and it responds to largely unidentified environmental signals. This review is focused on providing a global picture of virulence gene regulation in P. aeruginosa. In addition to key regulatory pathways that control the transition from acute to chronic infection phenotypes, some regulators have been identified that modulate multiple virulence mechanisms. Despite of a propensity for chaotic behaviour, no chaotic motifs were readily observed in the P. aeruginosa virulence regulatory network. Having a ‘birds-eye’ view of the regulatory cascades provides the forum opportunities to pose questions, formulate hypotheses and evaluate theories in elucidating P. aeruginosa pathogenesis. Understanding the mechanisms involved in making P. aeruginosa a successful pathogen is essential in helping devise control strategies.

INTRODUCTION

Pseudomonas aeruginosa is a Gram-negative bacterium that has the ability to thrive in most natural and man-made environments. It is found in diverse habitats, including soil, water, plants and animals, and can infect multiple hosts (1,2). Pseudomonas aeruginosa causes a wide variety of acute (short duration, typically severe) and chronic (persisting for a long time, often refractory to treatment, severity varying with pathogen) human infections, including in patients with severe burn wounds, urinary tract infections, AIDS, lung cancer, chronic obstructive pulmonary disease, bronchiectasis and cystic fibrosis (CF) (3–6).

Metabolic versatility, intrinsic and acquired antibiotic resistance, biofilm formation and production of multiple virulence (disease-causing) factors make P. aeruginosa a formidable pathogen. The virulence machinery of P. aeruginosa comprises both cell-associated determinants (such as lipopolysaccharides, pili, flagella) and numerous secreted factors (such as elastases, proteases, exotoxins, pyocyanin, extracellular polysaccharides). One of the mechanisms by which P. aeruginosa senses external signals is using sensor proteins that, through phosphotransfer or phosphorelay, activate specific transcriptional regulators. These sensor–regulator protein pairs are called two-component systems (TCS). The P. aeruginosa PAO1 genome encodes ∼127 TCS members, compared with 60 in Escherichia coli (7) and 70 in Bacillus subtilis (8), reflecting the adaptability of P. aeruginosa. TCS and their modifications also feed into major regulatory pathways and play a critical role in allowing cells to modulate gene expression in response to environmental conditions (9,10). Many of the secreted virulence factors and phenotypes, such as biofilm formation, are under the control of a cell density recognition mechanism called quorum sensing (QS) that aids in the coordinated expression of genes (11,12). QS is a key to virulence gene expression in many bacteria and serves as an attractive target for antibacterial chemotherapy (13).

In humans, acute P. aeruginosa infections in specific sites, such as the CF lung, eventually lead to chronic inections. This is caused by adaptive modifications in the infecting clonal type, resulting in diverse morphotypes (14). Acute virulence factors include the Type 2 and Type 3 secretion systems, flagella, type IV pili and QS-regulated virulence factors (proteases, elastase, pyocyanin) (15). On establishing a chronic infection, P. aeruginosa overproduces extracellular polysaccharides, forms biofilms and small colony variants and upregulates the Type 6 secretion system (15–18). Antibiotic resistance plays a major role in both types of infection, although the cells display higher levels of resistance in chronic infections (18,19). The transition to a chronic infection phase is the result of numerous changes in cellular physiology in response to external stimuli (20). The changes include downregulation of acute virulence genes with a concomitant upregulation of chronic infection phenotypes and antibiotic resistance, facilitating recalcitrant infections (15,20). Host invasion, establishment of acute infection and the subsequent transition to the chronic phase involves tightly regulated expression of many genes associated with metabolism, virulence and antibiotic resistance. Several key players in these transition processes have been identified and include transcriptional and post-transcriptional regulators (21–23). Many of these will be discussed in subsequent sections of this review.

Gene regulation in P. aeruginosa is a complex process involving numerous transcriptional regulators, regulatory RNAs (rgRNA) and σ factors. The P. aeruginosa genome is >6 Mb (24), approaching that of lower eukaryotes. The genome is plastic and has acquired genes and undergone extensive rearrangements to adapt to specific niches (25). The large genome of P. aeruginosa supports a multitude of regulatory networks, with ∼8% of the total genome dedicated to the regulatory proteins (26). Pseudomonas aeruginosa PAO1 encodes 434 transcriptional regulators, 24 σ factors and 34 small RNAs, many of which remain to be characterized (24,27–29). Moreover, predicted regulatory networks indicate that there is an extensive crosstalk between the different transcriptional regulators (27,30). These networks, however, are based in part on in silico analyses, and their validity needs to be established. This review makes an effort to consolidate the empirically proven major virulence regulatory networks in P. aeruginosa with the hope of providing a framework for future studies to better understand pathogenic processes in P. aeruginosa and in related bacteria.

MAJOR VIRULENCE REGULATORY SYSTEMS IN P. AERUGINOSA

The experimentally established virulence regulatory network in P. aeruginosa is depicted in Figure 1. Our group and others have previously performed in silico analyses of the P. aeruginosa transcriptional regulatory network (27,30,31). Comparing those networks with the network depicted in Figure 1 clearly demonstrates the gap in knowledge between predicted networks and established ones. An important contributing factor to this discrepancy is the fact that the functions of the majority of the genes in the PAO1 genome remain unknown. Deep sequencing, transcriptome metaanalysis (32,33) and complementary studies will aid in assigning functions to the hypothetical genes and undoubtedly narrow this knowledge gap.

Figure 1.

The P. aeruginosa virulence regulatory network. The pathogenic potential of P. aeruginosa is dictated by multiple virulence systems that are regulated transcriptionally, post-transcriptionally and post-translationally. The central mechanism for P. aeruginosa virulence regulation is QS, which controls expression of many virulence factors in a population density-dependent manner. Key activators of this system are LasR, RhlR, MvfR, VqsR, the cAMP receptor protein Vfr and the stationary phase σ factor RpoS. Las system repressors include RsaL, the H-NS protein MvaT, the σ factor RpoN and the sRNA-binding protein RsmA, whereas others like QscR repress both the Las and Rhl systems. Other regulators such as AmpR affect QS genes by an unknown mechanism. QS plays a role in regulating critical pathogenic mechanisms, including biofilm formation, secretion systems, production of numerous virulence factors, efflux pumps, antibiotic resistance and motility. Acute P. aeruginosa infections can lead to chronic infections in response to largely unidentified signals. A key regulatory pathway that controls this lifestyle switch is the RetS–LadS–GacSA–RsmA pathway. RetS and LadS are hybrid sensor proteins that, in response to external signals, either activate or repress the GacSA TCS. The GacA regulator then activates transcription of two rgRNAs, rgRsmZ and rgRsmY that sequester and inhibit activity of the sRNA-binding protein, RsmA. RsmA is a key activator/repressor that post-transcriptionally regulates numerous acute and chronic infection phenotypes, including multiple QS-regulated virulence factors, biofilm formation, Type 2, Type 3 and Type 6 secretion systems and motility. Another major phenotypic change associated with the switch from acute to chronic phases of infection is the formation of biofilm. This is associated with extensive changes in transcription. Three key TCS involved in activating biofilm formation are BfiSR, MifR and BfmSR. Cyclic-di-GMP is another major player influencing this process, whose levels are controlled by diguanylate cyclases and phosphodiesterases. QS and the cup genes enhance biofilms, whereas regulators like AmpR repress it. An important component of P. aeruginosa biofilms are extracellular polysaccharides, such as alginate, Pel and Psl. Alginate production is under the control of the master regulator ECF AlgT/U, whose activity is regulated transcriptionally by AmpR, post-translationally by MucA and MucB and by regulated intermembrane proteolysis involving MucP, AlgW, ClpXP and others. AlgT/U activates the alginate biosynthetic operon through AlgR, AlgB and AmrZ. In addition, biofilm formation is also affected by iron concentration, a process governed by the master repressor of iron uptake, Fur. Fur controls uptake of iron by regulating the σ factor PvdS, thereby modulating sidephore levels. Fur also modulates transcription of two key regulatory RNAs, asPrrF1 and asPrrF2. These two sRNAs are involved not only in regulating iron-uptake-related genes but also enzymes of the trichloroacetic acid cycle and genes involved in anthranilate synthesis. Anthralinate, a precursor for synthesis of PQS, is a key regulatory molecule of the PQS signalling system in P. aeruginosa, which is involved in expression of QS-regulated virulence factors. Details on the individual interactions and the appropriate references can be found in the text. Some of the interactions labelled as indirect are regulated by unknown mechanisms and warrant further investigation. In the figure, some regulators and phenotypes have been mentioned more than once.

Cis regulatory elements (CREs) form a critical part of transcription. CREs are non-coding DNA sequences present in or near a gene, and they often contain binding sites for transcription factors and/or other regulators of transcription (34). The two major CREs are promoters and enhancers (35,36). The promoters contain the binding sites for transcription factors and other regulatory molecules, such as σ factors and regulatory RNAs (37–39). Enhancers, once thought to be part of only eukaryotes, are found widely in prokaryotes also, and they function in conjunction with the σ54-RNA polymerase (40–42). The known P. aeruginosa transcription factor binding sites are listed in Table 1. This section will focus on the transcriptional and post-transcriptional regulation of critical pathways that determine P. aeruginosa pathogenesis.

Table 1.

Cis regulatory elements in P. aeruginosa transcriptional regulation

Transcription factorCis regulatory elementMajor virulence phenotype regulatedReference
AlgRACCGTTCGTCAlginate production, biofilm formation, T3SS(43)
AlgZGGCCATTACCAGCCAlginate production(44)
AnrTTGATN4ATCAAAnaerobic regulator of QS(45)
ArgRTGTCGCN8AACarbon and nitrogen catabolism(46)
ExsATNAAAANAT3SS(47)
FleQBox 1: CGCCTAAAAATTGACAGTTMotility, biofilm formation(48)
Box 2: CATTAGATTGACGTTAATC
FurGATAATGATAATCATTATCIron uptake(49)
LasR (las box)NHCTRNSNNDHNDKNNAGNBQS(50)
MexTATCAN5GTCGATN4ACYATAntibiotic resistance, T3SS, QS(51)
MvfRTTCGGACTCCGAAQS(52)
PsrAG/CAAACN2-4GTTTG/CStress regulon (RpoS), T3SS(53)
RcsBaTTA-GAAACGTCCTAAAFimbriae(54)
RhlR (lux box)CCTGTGAAT/ATCC/TGGT/CAGTTQS(55)
VfrAATTGACTAATCGTTCACATTTGQS(56)
VqsRTCGCCN8GGCGAQS(57)
Transcription factorCis regulatory elementMajor virulence phenotype regulatedReference
AlgRACCGTTCGTCAlginate production, biofilm formation, T3SS(43)
AlgZGGCCATTACCAGCCAlginate production(44)
AnrTTGATN4ATCAAAnaerobic regulator of QS(45)
ArgRTGTCGCN8AACarbon and nitrogen catabolism(46)
ExsATNAAAANAT3SS(47)
FleQBox 1: CGCCTAAAAATTGACAGTTMotility, biofilm formation(48)
Box 2: CATTAGATTGACGTTAATC
FurGATAATGATAATCATTATCIron uptake(49)
LasR (las box)NHCTRNSNNDHNDKNNAGNBQS(50)
MexTATCAN5GTCGATN4ACYATAntibiotic resistance, T3SS, QS(51)
MvfRTTCGGACTCCGAAQS(52)
PsrAG/CAAACN2-4GTTTG/CStress regulon (RpoS), T3SS(53)
RcsBaTTA-GAAACGTCCTAAAFimbriae(54)
RhlR (lux box)CCTGTGAAT/ATCC/TGGT/CAGTTQS(55)
VfrAATTGACTAATCGTTCACATTTGQS(56)
VqsRTCGCCN8GGCGAQS(57)

H = C/T/A; R = A/G; S = C/G; D = G/A/T; K = G/T; B = C/G/T; N = A/C/G/T.

aRcsB is found only in P. aeruginosa PA14, not in P. aeruginosa PAO1.

Table 1.

Cis regulatory elements in P. aeruginosa transcriptional regulation

Transcription factorCis regulatory elementMajor virulence phenotype regulatedReference
AlgRACCGTTCGTCAlginate production, biofilm formation, T3SS(43)
AlgZGGCCATTACCAGCCAlginate production(44)
AnrTTGATN4ATCAAAnaerobic regulator of QS(45)
ArgRTGTCGCN8AACarbon and nitrogen catabolism(46)
ExsATNAAAANAT3SS(47)
FleQBox 1: CGCCTAAAAATTGACAGTTMotility, biofilm formation(48)
Box 2: CATTAGATTGACGTTAATC
FurGATAATGATAATCATTATCIron uptake(49)
LasR (las box)NHCTRNSNNDHNDKNNAGNBQS(50)
MexTATCAN5GTCGATN4ACYATAntibiotic resistance, T3SS, QS(51)
MvfRTTCGGACTCCGAAQS(52)
PsrAG/CAAACN2-4GTTTG/CStress regulon (RpoS), T3SS(53)
RcsBaTTA-GAAACGTCCTAAAFimbriae(54)
RhlR (lux box)CCTGTGAAT/ATCC/TGGT/CAGTTQS(55)
VfrAATTGACTAATCGTTCACATTTGQS(56)
VqsRTCGCCN8GGCGAQS(57)
Transcription factorCis regulatory elementMajor virulence phenotype regulatedReference
AlgRACCGTTCGTCAlginate production, biofilm formation, T3SS(43)
AlgZGGCCATTACCAGCCAlginate production(44)
AnrTTGATN4ATCAAAnaerobic regulator of QS(45)
ArgRTGTCGCN8AACarbon and nitrogen catabolism(46)
ExsATNAAAANAT3SS(47)
FleQBox 1: CGCCTAAAAATTGACAGTTMotility, biofilm formation(48)
Box 2: CATTAGATTGACGTTAATC
FurGATAATGATAATCATTATCIron uptake(49)
LasR (las box)NHCTRNSNNDHNDKNNAGNBQS(50)
MexTATCAN5GTCGATN4ACYATAntibiotic resistance, T3SS, QS(51)
MvfRTTCGGACTCCGAAQS(52)
PsrAG/CAAACN2-4GTTTG/CStress regulon (RpoS), T3SS(53)
RcsBaTTA-GAAACGTCCTAAAFimbriae(54)
RhlR (lux box)CCTGTGAAT/ATCC/TGGT/CAGTTQS(55)
VfrAATTGACTAATCGTTCACATTTGQS(56)
VqsRTCGCCN8GGCGAQS(57)

H = C/T/A; R = A/G; S = C/G; D = G/A/T; K = G/T; B = C/G/T; N = A/C/G/T.

aRcsB is found only in P. aeruginosa PA14, not in P. aeruginosa PAO1.

QS

QS is a signalling mechanism that bacteria use to regulate gene expression in a population density-dependent manner, and it was first demonstrated in Vibrio fischeri (58). In QS, the bacteria produce and secrete small molecules called autoinducers or quoromones. When these molecules reach a concentration threshold, they diffuse back into the cell to elicit a coordinated response promoting group survival (59). Pseudomonas aeruginosa uses QS to regulate production of various virulence determinants, such as extracellular proteases, iron chelators, efflux pump expression, biofilm formation, motility and the response to host immune signals (60). This is achieved using two types of autoinducers, N-acyl-homoserine lactones (AHLs) and 2-alkyl-4 quinolones (AQs) (61).

AHL-mediated QS

Pseudomonas aeruginosa has two canonical AHL QS signalling pathways, the las and rhl systems. Together, these pathways affect expression of ∼10% of the P. aeruginosa transcriptome (62). The lasI (PA1432) and rhlI (PA3476) genes encode the N-3-oxododecanoylhomoserine lactone (3-oxo-C12-AHL) synthetase (63,64) and N-butyrylhomoserine lactone (C4-AHL) synthetase, respectively (65–68). The resulting AHLs then bind and activate their cognate LuxR family regulators, LasR (PA1430) (64) or RhlR (PA3477) (67). LasR and RhlR multimerize in the presence of their cognate AHL (69,70). In in vitro studies, LasR–DNA interaction is cooperative and non-cooperative in the presence or absence of a dyad symmetry in the binding sites, respectively (71). Rhl-regulated promoters have binding sites with a dyad symmetry (72).

AQ-mediated QS

Pseudomonas aeruginosa synthesizes two AQ QS signals, 2-heptyl-3-hydroxy-4-quinolone (PQS) and its precursor, 2-heptyl-4-quinolone (HHQ) (73). Both PQS and HHQ enhance in vitro binding of the LysR-type transcription regulator, MvfR (also known as PqsR, PA1003), to the promoter of the pqsABCDE operon (PA0996–PA1000), suggesting that they function as MvfR effectors (74). Microarray analysis identified 141 genes differentially expressed in an mvfR mutant strain, including lasR, algT/U (PA0762), rsmA (PA0905) and rsaL (PA1431) (75). PQS also acts independently of MvfR to induce expression of the Fur regulon through its ability to bind iron (73,76) and membrane vesicle formation by inducing membrane curvature (77,78). PmpR (PA0964), a YebC member, negatively regulates MvfR (Figure 1) (79).

QS regulation

The las, rhl and PQS/HHQ/MvfR systems exhibit positive feed-forward autoregulation (52,80). In addition, the P. aeruginosa AHL systems function in a hierarchical manner, as the 3-oxo-C12-AHL-LasR complex positively regulates rhlI, rhlR and mvfR expression as well as lasI (81–83). Exceptions to this have been noted. RhlR has been shown to regulate LasR-dependent genes in strains lacking lasR (84), and timing of lasI, lasR, rhlI and rhlR expression can vary drastically depending on growth conditions (85).

Many global regulators have been shown to modulate QS-dependent genes. RpoS (PA3622), the stationary phase σ factor affects ∼40% of the QS regulon (72,86). RpoS binding sites have been identified in several of the QS-dependent promoters. RpoS also affects lasR and rhlR expression, and LasR binding sites have been identified in promoters of other transcriptional regulators in the QS regulon, including PA2588, PA4778, pvdS (PA2426), vqsR (PA2591) and rsaL (87). Chromatin immunoprecipitation studies have shown occupancy by histone-like silencers MvaT (PA4315) and MvaU (PA2667) on lasI, lasR, mvfR, rpoS and rsaL (88). RsaL plays an important role in las signalling homeostasis, by binding to the lasI promoter and preventing LasR-mediated activation (89). In addition to affecting gene expression through las regulation, microarray analyses indicate that RsaL affects expression of 130 genes, including direct regulation of pyocyanin and hydrogen cyanide genes (89). RsaL also seems to be important in regulating the transition from planktonic to a sessile state, as rsaL mutants exhibit increased swarming motility and fail to form biofilms (90). RsaL expression is under the control of LysR-type regulator OxyR (PA5344) (91). The lasI promoter region has also been shown to be bound by CzcR (PA2523), which is required for expression of rhlI and rhlR in addition to lasI (Figure 1) (92). CzcR is part of the CzcRS TCS, which is shown to be involved in carbapenem and heavy metal resistance (93).

VqsR (PA2591), which is induced by H2O2 or human serum (94) and is under LasR regulation (95), regulates QS through inhibition of the LuxR-type regulator, QscR (PA1898, Figure 1) (96). Although QscR binds to 3-oxo-C12-AHL, its specificity is not as stringent as LasR (97). The QscR regulon partially overlaps that ascribed to the las and rhl systems, but also has unique targets (98). In the absence of AHL, QscR can multimerize and form heterodimers with LasR and RhlR (99). QscR also plays a role in LasI homeostasis, as mutations in qscR result in premature lasI expression (100). An AraC family member VqsM (PA2227) regulates VqsR in addition to numerous genes involved in QS, including RsaL, PprB (PA4296), MvfR, RpoS as well as AlgT/U and MexR (PA0424) (101).

Additionally, pqsH (PA2587), which encodes the enzyme responsible for oxidation of HHQ to form PQS, is positively regulated by the las system (102,103) and is negatively regulated by the rhl system (52). PQS is derived from anthranilate, which is synthesized by the kynurenine pathway (104). Kynurenine pathway anthranilate is also required for N-decanoyl-homoserine lactone (C10-AHL) dependent signalling, which is independent of las, rhl and qscR (104). The receptor for this signalling is yet to be identified (105). Besides potential heterodimerization with QscR, additional post-transcriptional regulation of QS has been described. In one such mechanism, QteE (PA2593) destabilizes LasR and RhlR, and in the absence of qteE, the quorum threshold-requirement for activation of QS-dependent genes is lost (106). RsmA negatively regulates rhl and las signalling, resulting in reduced AHL levels (107). Moreover, it has been shown the RNA chaperone Hfq (PA4944) positively regulates rhlI translation through rsmY and RsmA (108).

Recently, our laboratory has established a role for the ß-lactamase regulator AmpR (PA4109) in activating QS-regulated genes (23). The production of QS-regulated secreted virulence factors, such as LasA (PA1871) and LasB (PA3724) proteases, and pyocyanin production is significantly impaired in AmpR-deficient strains. Further, loss of ampR reduced virulence in the Caenorhabditis elegans toxicity assay (23,109). In addition, AmpR regulates non–ß-lactam resistance by repressing activity of the MexEF–OprN (PA2493–PA2495) efflux pump, the alginate master regulator AlgT/U (110) and biofilm formation (23), suggesting that it plays a role in maintaining the acute mode of infection.

TCS

TCS are sophisticated signalling mechanisms marked by a highly modular design that have been adapted and integrated into a wide variety of cellular signalling circuits. The archetypical TCS is composed of a membrane integrated sensory histidine kinase (HK) and a cytoplasmic response regulator (RR) (111). The HK contains a periplasmic N-terminal domain that detects specific stimuli (sensing domain) and a C-terminal cytoplasmic transmitter domain that comprises a dimerization domain, a conserved histidine and an adenosine triphosphate catalytic domain (112). HKs can have two or more transmembrane domains with little or no periplasmic domain, whereas others are completely cytoplasmic. The cognate RR contains a conserved receiver domain and a variable output domain (113). On receiving a signal, two HK monomers dimerize and cross-phosphorylate at the conserved histidine residue, and the phosphate is subsequently transferred to an aspartate residue in the receiver domain of the cognate RR (114). The phosphotransfer is catalysed by the receiver domain, and it results in a conformational change that activates the output domain, which often binds DNA and modulates gene expression or enzymatic activity (9,113,115). Variations to this model occur in phosphorelays, where a sensor kinase first transfers the phosphoryl group to an RR that has no output domain. This P∼RR then transfers the phosphoryl group to a histidine-containing phosphotransfer protein, and this in turn serves as a phosphate donor to a terminal RR, which has an output domain mediating a cellular response (10). In other cases, the sensor kinase and the RR lacking an output domain are fused into one protein (hybrid sensor kinase) (116). Other variations include the TCS connectors, a group of proteins that modulate the phosphorylation state and activity of sensor HK and RR and establish regulatory links between otherwise independent signal transduction pathways (117).

Pseudomonas aeruginosa, equipped with 55 HKs, 89 RRs and 14 HK–RR hybrids, possesses one of the largest pool of TCS proteins identified in any microorganism analysed thus far (24). This provides the bacterium with a sophisticated capability to regulate diverse metabolic adaptations, virulence and antibiotic resistance processes that are hallmark of P. aeruginosa infections. One of the critical TCSs is GacSA (GacS-PA0928, GacA-PA2586), which is central to expression of virulence factors, secondary metabolites, biofilm formation and QS (107,118) and is the switch between acute and chronic infections (1,119). GacS is a hybrid sensor HK that contains an HK domain, an RR domain and a histidine phosphotransfer (Hpt) domain (21,120). GacS phosphorylation is under the control of two hybrid sensor kinases, RetS (PA4856) (21) and LadS (PA3974) (22) (Figure 1). RetS can directly interact with GacS and prevent GacS phosphorylation (22,121), whereas LadS phosphorylates GacS (22). Phosphorylated GacA positively regulates the transcription of two small regulatory RNAs, rgRsmZ (PA3621.1) and rgRsmY (PA0527.1), which block the negative regulator RNA-binding protein RsmA (PA0905). RsmA positively regulates genes of the Type 3 secretion system, type IV pili formation and iron homeostasis while repressing QS, Type 6 secretion and potentially other transcription factors (122–124). The GacSA TCS is also involved in antibiotic resistance to three different families of antibiotics, tobramycin, ciprofloxacin and tetracycline (125), apparently through RsmA/rgRsmZ.

In P. aeruginosa, PhoPQ (PA1179–PA1180) together with PmrAB (PA4776–PA4777) are two TCSs that respond to limiting concentrations of cations, and regulate resistance to polymyxin B and cationic antimicrobial peptides through the regulation of the arnBCADTEF-pmrE (PA3552PA3559) LPS modification operon (126,127). PhoQ is involved in swarming and twitching motility as well as in biofilm formation and is required for virulence without affecting the T3SS or QS systems (Figure 1) (128). The HK PhoQ activates the RR PmrA independently of PmrB, suggesting an interaction between these TCSs (129). In addition, increased resistance to antibiotics, including polymyxin B, aminoglycosides and quinolones in phoQ mutants suggests crosstalk between PhoPQ and other TCSs (130,131).

Formation of biofilms

Biofilms are surface-associated multicellular bacterial communities encapsulated in a self-produced extracellular matrix composed of polysaccharides, proteins and nucleic acids that mediate cell-to-cell and cell-to-surface interactions (132). Pseudomonas aeruginosa biofilms, typically associated with poor patient prognosis, signify the switch from an acute to a chronic infection. Biofilms can be formed on abiotic (environment) or biotic (wounds, surgical implants, CF lung) surfaces (133). Biofilm formation and maintenance is tightly regulated in response to environmental cues, conferring enhanced resistance against antimicrobial agents and immune defence mechanisms on the biofilm bacteria (12). Formation of biofilms is a multi-stage process that is initiated by the surface attachment of planktonic bacteria to form a monolayer, clonal growth/aggregation leading to the formation of microcolonies, maturation to form mushroom-shaped structures and dispersal (134–136). As can be imagined, this complex transition in the bacterial lifestyle is accompanied by drastic changes in gene regulation.

Surface attachment by P. aeruginosa to form microcolonies has been attributed to type IV pili, flagella, free DNA, alginate and Pel and Psl polysaccharides, although pili, alginate and flagella mutants also form biofilms (136,137). Attachment is a reversible process, and the commitment to form biofilms is partly under positive SadB (PA5346) regulation (138). SadB upregulates both Pel polysaccharide production and the chemotaxis-like cluster CheIV (PA0408–PA0417), which is thought to regulate flagellar motion by an unknown mechanism (139).

The Cup fimbriae, encoded by three distinct gene clusters cupA (PA2128PA2133), cupB (PA4081PA4086) and cupC (PA0992, PA0993, PA0994) in P. aeruginosa PAO1, have been demonstrated to play a role in different stages of biofilm formation on biotic and abiotic surfaces (140). Regulation of the cup genes is complex, involving a phase variation-dependent repression of cupA expression by an H-NS member MvaT (141,142). MvaT also regulates the cupB and cupC loci to a lesser extent (141). The cupB and cupC clusters are under the primary regulation of the RocS1–RocR–RocA1 (PA3946–PA3948) three-component system (143). This system is similar to the Bordetella pertussis BvgASR system (144) and consists of the hybrid sensor kinase RocS1, the response regulator RocA1 and the RocA1-repressor RocR (143,145). RocR has been hypothesized to bind to c-di-GMP through its EAL (diguanylate phosphodiestrerase) domain and prevents phosphotransfer from RocS1 to RocA1, thus preventing RocA1 activation (143,145). Pseudomonas aeruginosa PA14 has a fourth cup cluster (cupD) on the pathogenicity island PAPI-I, which is controlled positively by the response regulator RcsB (PA4080) and negatively by the EAL-domain containing response regulator PvrR (146). In addition, diguanylate cyclases and phosphodiesterases of the wsp gene cluster (PA3702–PA3708) (147,148) MorA (PA4601) (149) and TpbA–TpbB (PA3885, PA1120) (150) modulate intracellular levels of c-di-GMP to exert a regulatory effect on the cup gene clusters (Figure 1).

The main components of the extracellular polymeric substance matrix of biofilms are Pel and Psl polysaccharides, alginate and free DNA (12,136). Both pel and psl gene loci are post-transcriptionally regulated by the RetS–LadS (PA4856 and PA3974, respectively) system through rsmY and rzmZ (21,22,121) and by c-di-GMP levels, either directly (147) or by binding the transcriptional regulator FleQ (PA1097) (151). The pel operon is also repressed by the las QS system through the tyrosine phosphatase TpbA (PA3885) (150). A membrane-bound sensor, PpyR (PA2663) enhances biofilm formation through the psl operon and virulence through modulating QS (152). Although alginate is a major component of biofilms and affects biofilm structure, it is not essential for biofilm formation (136). Alginate regulation is discussed in a separate section (see later in the text). QS regulates cell lysis in biofilms (153–155), thereby controlling the release of extracellular DNA, a major component of the biofilm matrix (156,157). The QS system also regulates rhamnolipid production (67) that promotes motility and, hence, formation of the cap in the mushroom structure of mature biofilms (158), and maintenance of biofilm channels (159). BfiRS (PA4196–PA4197), BfmRS (PA4101–PA4102) and MifR (PA5511) are TCSs shown to regulate biofilm development and maturation by sequential phosphorylation (160). They activate biofilm formation at different transition stages, reversible to irreversible attachment (BfiRS), irreversible attachment to maturation stage-1 (BfmRS) and maturation stage-1 to mushroom structure formation (MifR) (Figure 1) (160). The BfiRS system may function in conjunction with the GacSA TCS and feed into the Rsm loop of regulation to control biofilm formation (160). SagS (PA2824), the cognate sensor of HptB (PA3345), modulates biofilm development (by controlling BifS phosphorylation) and other virulence phenotypes (by modulating rgsRmZ levels) depending on whether the cells are in the planktonic or biofilm phase (161).

Analyses of clinical isolates reveal a positive correlation between expression of lasR, rhlR and acute virulence factors (162,163), suggesting that QS is required for virulence in vivo. QS is also important when P. aeruginosa grows as biofilms in the CF lung (164). In vivo studies show that lasI and rhlI mutants produce milder chronic lung infections compared with their wild-type counterparts (165) and form more susceptible biofilms (166). However, in some in vitro studies, there was no apparent difference in the biofilms formed by the QS mutants and the wild-type strains (167,168). This discrepancy in the requirement of QS for biofilm formation and establishment of a successful chronic infection is probably not surprising, as QS regulates many different functions. Further, it has been demonstrated that the CF environment selects for strains with lasR mutations, although the rhl system is intact (169). Although lasR is higher up in the QS hierarchy, studies have shown that secondary mutations can re-establish rhl expression in las mutants (170). This suggests that in CF biofilms, the rhl system is more important, and lasR inactivation serves to downregulate the acute virulence factors (171).

A major cause of antibiotic resistance in biofilms has recently been attributed to the phenomenon of persistence. Persister cells are small subpopulations of antibiotic-sensitive cells that have acquired transient antibiotic tolerance (172). When the antibiotic levels drop, the persisters grow into a population of sensitive cells, again with a small sub-population of persisters (173,174). Many genes involved in the formation of persisters have been identified in P. aeruginosa PA14, including two transcriptional regulators AlgR (PA5261) and PilH (PA0409) (175). However, this topic is outside the scope of this review but has been extensively reviewed elsewhere (176,177).

Alginate production

In the lungs, especially in patients with CF, P. aeruginosa can convert from a non-mucoid to an alginate-overproducing mucoid phenotype signalling chronic infection (178). Chronic P. aeruginosa infection seems to be localized to foci within the anaerobic mucus environment in the lung’s respiratory zone (179–182). These foci lead to tissue damage decreasing lung function, and the appearance of the mucoid phenotype correlates with poor patient prognosis (183,184). The exopolysaccharide alginate is a linear polymer of β-d-mannuronic acid and α-l-guluronic acid (185), which stimulates production of IgG and IgA antibodies (186). Although production of alginate is metabolically taxing, it protects the bacteria from phagocytosis and antibodies, thus conferring a survival advantage (187,188). Conversion to mucoidy occurs when biofilms are treated with activated polymorphonuclear leucocytes (189), hydrogen peroxide (189), antibiotics (190) and nutrient starvation (191,192).

A complex regulatory pathway controls alginate biosynthesis. The central player is the σE family extracytoplasmic function σ factor AlgT/U (PA0762) (193,194), whose activity is inhibited post-transcriptionally by the anti-σ factor MucA (PA0763) and by MucB (PA0764) (195–197). Loss of function mutations in mucA or mucB result in a mucoid phenotype (195,198,199) because of release of AlgT/U from MucA by a regulated intramembrane proteolytic pathway [reviewed in (200,201)]. It was recently demonstrated that MucA proteolysis is regulated not only by AlgW (PA4446) but also by MucD (PA0766) by activating the MucP protease (PA3649) (202). In addition, AmpR links alginate production with antibiotic resistance and QS by negatively regulating algT/U expression (Figure 1) (110). AlgT/U regulates alginate production at least in part by autoregulation (193), controlling expression of the transcriptional regulators algR (203,204), algB (198,204), amrZ (205) and the algD (PA3540) alginate biosynthetic operon (204,206,207). AlgB (208), AlgR (43,209,210) and AmrZ (211) directly bind to the algD operon to activate transcription. The alternative σ factor RpoN (PA4462) is also required for high levels of algT/U and algD expression (212).

The algB (PA5483) and algR (PA5261) genes encode NtrC and LytR subfamily, respectively, of TCS RRs (213). Interestingly, aspartic acid phosphorylation in the regulatory domain is not essential for alginate production (214). Transcriptome analysis of a PAOmucA22 mucoid strain (PDO300) (196) identified seven predicted transcriptional regulators, PA1235, PA1261, PA1637 (KdpE), PA2881, PA3420, PA3771 and PA5431, and one sensor kinase, EraS (PA1979), whose expression was downregulated in an algB mutant but not in a strain containing a mutation in its cognate TCS sensor, KinB (PA5484) (208). In addition to regulating the algD operon, AlgR directly activates transcription of algC (PA5322), which encodes a phosphomannomutase/phosphoglucomutase essential for Psl, alginate and rhamnolipid synthesis (Figure 1) (215–218). AlgR also is important for mature biofilm formation, possibly by directly repressing rhl-QS (219), type IV pilus formation by binding to the fimTUpilVWXY1Y2E promoter (220,221) and hydrogen cyanide production by binding to the hcnA (PA2193) promoter (222). Interestingly, in contrast to alginate production, the phosphorylation site is required for regulating cyanide production and twitching motility (220,223). AlgR has also been shown to indirectly regulate the cyclic AMP/Vfr-dependent pathway (224). The AlgR regulon has been characterized by several transcriptome studies (219,222,225). Two other regulators of alginate production in P. aeruginosa are Alg44 (PA3542) (226) and a diguanylate cyclase, MucR (PA1727) (227). MucR produces a pool of c-di-GMP in the vicinity of the PilZ domain of Alg44 (PA3542), which then positively regulates alginate production (Figure 1) (226–228).

Regulation of iron uptake

Iron is critical for growth of all organisms, and P. aeruginosa is no exception. Transcriptome studies reveal that a large number of genes are regulated in response to iron (229,230). Biologically useful iron (Fe2+) in the environment is scarce and is available mostly in the insoluble Fe3+ form. To help scavenge this free iron, bacteria produce siderophores that bind extracellular iron and transport them back into the cell through TonB-dependent receptors on the cell surface (231). Pseudomonas aeruginosa produces two siderophores, pyoverdine and pyochelin, and can also subvert siderophores produced by other organisms to take up haem (232,233). However, excess free iron in the cell leads to formation of toxic reactive oxygen species, and, therefore, cells tightly regulate the uptake (234). The ferric uptake regulator (Fur, PA4769) is a conserved protein in P. aeruginosa and other Gram-negative bacteria, and it is a major iron acquisition regulator (235). Fur dimerizes rapidly after synthesis, and it takes a minimum of two dimers to bind promoters of genes under Fur regulation in P. aeruginosa (236). Fur controls the iron regulon directly by binding the Fur box (237) and indirectly by modulating expression of other regulators, including the pyochelin uptake regulator PchR (PA4227), ECF σ factors like PvdS, TCS regulators and small regulatory RNAs [asPrrF1 (PA4704.1), asPrrF2 (PA4704.2); Figure 1] (237–239).

Iron concentrations in the cell also influence expression of virulence factors in P. aeruginosa. PvdS, for example, is critical in linking iron and virulence by controlling the production of pyoverdine, an outer membrane pyoverdine receptor [FpvA (PA2398)] and two important extracellular virulence factors [PrpL (PA4175) and exotoxin A (PA1148); Figure 1] (240–242). Also, pvdS mutants showed reduced virulence in a rabbit endocarditis model (243). Human lactoferrin inhibits P. aeruginosa biofilm formation, indicating a role for iron in the process (244). Iron chelation by lactoferrin induces twitching motility in P. aeruginosa negating colonization and ultimately, biofilm formation (244,245). Intracellular iron concentrations are one of the signals for biofilm development in a process involving Fur but not the iron uptake regulatory RNAs asPrrF1 and asPrrF2 (246). Further, high levels of iron suppress the PQS system, release of extracellular DNA and biofilm formation (247).

The link between iron and QS systems in P. aeruginosa is complex. QS systems are enhanced under limiting iron concentrations (85,248,249), and major QS regulators are also involved in regulating iron responsive genes (75,250,251). MvfR, for example, has been demonstrated to control transcription of iron-related genes, and it has an iron-starvation box in its promoter, a site recognized by PvdS (PA2426) to turn on transcription under low-iron concentrations (229,252). It was recently demonstrated that iron levels affect activity of the MvfR signalling molecule HAQ, adding another layer of complexity to the role of iron in QS (253). Another major QS and virulence regulator, VqsR regulates phenazine production by modulating phnAB expression (94). Moreover, the small regulatory RNAs asPrrF1 and asPrrF2, which are negatively regulated by Fur, positively regulate PQS production (Figure 1) (254). PQS has been shown to accumulate in the outer membrane and in membrane vesicles (77). PQS chelates iron, and this facilitates pyochelin and pyoverdin in scavenging iron (73,76).

Thus, iron uptake regulation in P. aeruginosa is a complex affair and involves multiple regulators that affect expression of numerous genes either by themselves or through other regulators. In addition, the interconnections between iron uptake mechanisms and other virulence systems, such as QS and biofilm formation, demonstrate the versatility of this bacterium in being able to pragmatically read environmental signals to accordingly modulate gene expression.

Toxins and exoproteins

Exoproteins are an important component of bacterial survival not only because they allow the bacteria to interact with their immediate environment and other organisms in the vicinity but also because they play a critical role in virulence. P. aeruginosa has a large complement of secreted proteins and five (type I, II, III, V and VI) of the seven secretion systems characterized in bacteria (255). A majority of the secreted proteins are toxins that aid in P. aeruginosa virulence, most of which, including LasA, LasB, PrpL, ToxA and phospholipases [PlcH (PA0844), PlcN (PA3319), PlcB (PA0026)], is secreted through the Xcp type II secretion system (T2SS) (255). Effector molecules that are crucial for evading the host phagocytic response are secreted through a dedicated T3SS (256), whereas the type I system (T1SS) secretes the alkaline protease AprA (257,258) and the haemophore HasAp (PA3407) (259). Substrates of the recently identified T6SS are just being discovered (260). In addition, c-di-GMP levels, modulated by the diguanylate cyclase WspR (PA3702) is involved in the switch between T3SS and T6SS independent of RetS but is dependent on rgRsmY and rgRsmZ (Figure 1) (261).

HasAp, a T1SS-secreted haem-uptake protein in P. aeruginosa, is under QS control (250). QS is also known to regulate PrpL that targets the human lactoferrin (see previous section) (240). Thus, QS in P. aeruginosa not only regulates enzymes to degrade the human lactoferrin but also produces proteins to retrieve the iron from the degraded lactoferrin. The other known T1SS substrate, AprA, is regulated by a novel LTTR named BexR (PA2432) that controls bistability in P. aeruginosa (Figure 1) (262). Inactivation of QS has been demonstrated to reduce expression of T2SS-secreted proteases, chitinases and lipases (63,263) because of downregulation of the Xcp T2SS (264,265). The TCS PhoBR (PA5360–PA5361) regulates other T2SS-secreted exoproteins, such as PlcH, PlcC, PlcN and the Hxc T2SS secreted alkaline phosphatase LapA (266,267). Microarray analysis revealed that a novel cell-surface signalling system PUMA3 regulates Hxc T2SS genes (268). The three T6SS systems (HSI-I, HSI-II and HSI-III) in P. aeruginosa are differentially regulated by the QS systems (269). Although LasR and MvfR negatively regulate the HSI-I system, they positively regulate expression of the functionally redundant HSI-II and HSI-III (Figure 1) (269). In addition, a putative regulator Sfa3 (SfnR) in P. aeruginosa PA14 (an orthologue of PA2359 in PAO1) potentially regulates the HSI-III cluster (269). HSI-I expression is also regulated by RetS through RsmA (270).

Pseudomonas aeruginosa T3SS is regulated in a complex and multi-tiered process, and it is probably the most well understood (271). Expression of T3SS is regulated transcriptionally and post-transcriptionally in response to host cell contact and environmental Ca2+ levels (272,273). ExsA (PA1713), an AraC member, regulates expression of the 43 genes that form the T3SS in P. aeruginosa by binding as a monomer to an A-rich 8-bp region upstream of the −35 in the promoter of genes under its regulation (272,47). ExsA autoregulates its own expression and is also activated by PsrA (PA3006), a member of the TetR family (274,275). Two anti-activators [ExsD (PA1714) and PtrA (PA2808)] also regulate ExsA-mediated activation (Figure 1). T3SS transcription is coupled to secretion and involves the anti-activator ExsD and the anti–anti-activator ExsC (PA1710) that regulate ExsA function. Under non-inducing conditions (high Ca2+), ExsE (PA1711) binds the anti–anti-activator ExsC, allowing the anti-activator ExsD to bind ExsA and inhibit transcription. Under Ca2+ delimiting conditions, ExsE is secreted, freeing ExsC to bind ExsD. Free ExsA then activates T3SS expression (Figure 1) (276). Although this is the primary mode of control, T3SS can also be triggered by stress because of DNA damage (RecA-mediated activation of PtrB) (277), high salt (278,279), metabolic stress (123,279,280), alginate regulators AlgT/U, AlgR and MucA (281), the MexEF–OprN efflux pump regulator MexT (PA2492) through PtrC (282) and the RetS/LadS/Gac-Rsm TCSs (21,22,283). Under low oxygen conditions, the anaerobic regulator Anr activates the response regulator NarL (PA3879), which in turn represses rgRsmY and rgRsmZ expression, allowing RsmA to activate T3SS (284). Expression of T3SS, however, happens only in a subset of the population even under inducing conditions (279,285). Multiple levels of control allow fine-tuning of T3SS expression, allowing P. aeruginosa to sense various environmental conditions and regulate expression in conjunction with other virulence factors.

Regulatory RNAs in P. aeruginosa virulence

RNAs other than messenger RNAs, transfer RNAs or ribosomal RNAs are termed small RNAs (sRNAs), and they affect all steps in gene expression pathways in both prokaryotes and eukaryotes (286). In general, sRNA-mediated regulation occurs in one of two ways, base pairing with DNA or mRNA, or by affecting the activity of a protein or protein complex (286). Not surprisingly, virulence gene expression in P. aeruginosa also relies on small rgRNA-mediated post-transcriptional regulation. The importance of rgRNAs in regulation of bacterial virulence is well established (39,287,288). Most of the P. aeruginosa rgRNAs that have been characterized play a role in virulence gene regulation (discussed later in the text). A recent study identified ∼150 novel sRNAs using sRNA-Seq in P. aeruginosa PAO1 and PA14, which includes both strain-specific and shared ones (289).

Perhaps the most well characterized system involves the rgRNAs, rgRsmY and rgRsmZ (whose roles in virulence regulation have been discussed in the ‘TCS’ and ‘Toxins and Exoproteins’ sections). These are two functionally redundant rgRNAs in P. aeruginosa that play a critical role in the switch between acute and chronic infections (21,290). GacA of the GacSA TCS positively regulates expression of rgRsmY and rgRsmZ, which then bind to and sequester the sRNA-binding protein RsmA through the GGA motif (291–293), leading to derepression of the genes that RsmA represses (107,124,294,295). The consequences of RsmA sequestration result in dysregulation of the expression of numerous virulence factors, as discussed in the ‘TCS’ section. Given the importance of this regulatory process in P. aeruginosa pathogenesis, regulation of expression of rgRsmY and rgRsmZ is multi-tiered. The histidine phosphotransfer protein HptB is phosphorylated by a phosphorelay involving the three sensor kinases PA2824, PA1611 and PA1976 (296). Phosphorylated HptB then transfers the phosphate to an anti–anti-σ factor PA3374, to negatively regulate expression of rsmY (296,297). In another mode of regulation, the BfiSR TCS activates expression of the ribonuclease CafA (PA4477), which specifically targets rgRsmZ (298). Further regulation is achieved by the global regulators of the H-NS family of proteins MvaT and MvaU, which bind to AT-rich regions upstream of the rsmZ gene repressing their expression (299). In addition to all this, there is a negative autoregulatory feedback mechanism, the details of which have not been elucidated yet (300). On synthesis, rgRsmY is stabilized by Hfq binding, either alone or in conjunction with RsmA (108,301).

Another example of post-transcriptional regulation by sequestering a RNA-binding protein links virulence with metabolism. Pseudomonas aeruginosa Crc (PA5332) is a RNA-binding protein that recognizes CA-motifs around the ribosome binding sites of the mRNA of carbon compound catabolism genes. Crc thus represses genes whose products help utilize less preferred carbon sources (302–304). When less preferred substrates, such as mannitol, have to be utilized, expression of catabolic genes is achieved by sequestration of Crc by the rgRNA, rgCrcZ (305). Expression of rgCrcZ is under the control of the TCS CbrAB (PA4725–PA4726), which in conjunction with Crc plays a role in carbon compound catabolism, biofilm formation, antibiotic resistance, secretion systems and swarming (306–312).

Pseudomonas aeruginosa antisense sRNAs (asRNAs) can also act by base pairing with target mRNAs, thus inhibiting translation (313). One such example is asPhrS (PA3305.1), which plays a role in PQS and pyocyanin expression (314). Transcriptome studies indicate an extensive overlap between the genes that are positively regulated by the transcriptional regulator PqsR (also known as MvfR, PA1003) and asPhrS, suggesting that asPhrS regulates pqsR mRNA (75,314). Interestingly, it was shown that asPhrS specifically targets a region in the RBS of a small ORF (uof), which is present upstream of PqsR (314). As translation of pqsR and uof are coupled, asPhrS regulates pqsR translation by modulating translation of uof (314). Expression of asPhrS is under the control of the oxygen responsive regulator Anr (314). Hfq controls asPhrS expression indirectly by regulating Anr expression, whose mechanism of action is yet to be elucidated (314,315).

Small asRNAs also play a role in regulation of iron uptake and involve base pairing by the sRNAs asPrrF1 and asPrrF2, which are the P. aeruginosa orthologues of E. coli RyhB (237,316). Expression of asPrrF1 and asPrrF2 is repressed by Fur when iron concentrations are high (237). Under iron-starvation conditions, asPrrF1 and asPrrF2 are expressed and base pair with the mRNA of target genes, which include the superoxide dismutase sodB (PA4366), genes involved in the trichloroacetic acid cycle and anthranilate and cathechol degradation (317). Thus, asPrrF1 and asPrrF2 link carbon metabolism, iron uptake and QS-mediated virulence. Another asRNA gene asPrrH is located in the same locus as asPrrF1 and asPrrF2. The asPrrH asRNA (at 325 nt) is longer than asPrrF1 (116 nt) and asPrrF2 (114 nt), and the coding region of asPrrH overlaps with the asPrrF1 terminator, the intergenic region between asPrrF1 and asPrrF2 and the 5′-end of the asPrrF2 ORF (318). The expression of asPrrH is maximal in the stationary phase of growth, similar to asPrrF1 and asPrrF2, and under iron-deplete conditions (318). Haem represses asPrrH expression, and this involves the outer membrane haem receptors PhuR (PA4710) and HasR (PA3408) (318). Interestingly, under conditions of haem starvation, asPrrH expression leads to the repression of achAB and sdhCDAB, which are also targets of the PrrF asRNAs (318). In addition to these targets, asPrrH also represses NirL, a protein involved in biosynthesis of haem, under haem and iron limitation (318).

CONCLUSIONS AND PERSPECTIVES

Pseudomonas aeruginosa is a versatile bacterium that can thrive in a wide range of habitats. This is achieved by an intricately interlinked regulatory system of transcriptional regulators, σ factors, sRNAs and their regulons. The exquisite control of gene expression is exemplified in the virulence regulatory network (Figure 1), which demonstrates that none of the virulence mechanisms are isolated. Expression of individual virulence networks is under transcriptional and post-transcriptional regulation of multiple-regulatory systems, either directly or indirectly. Furthermore, some signalling cascades inversely regulate the acute and chronic virulence phenotypes depending on the signals sensed. The next critical phase of research should focus on the signals that the bacteria recognizes to achieve gene regulation.

The extent of cross-regulation between the transcriptional regulators highlights the global nature of the regulation, where individual subnetworks (such as the QS network, alginate network and so forth) are interlinked to form a hyperconnected network (Figure 1). Given the complexity of the connections, one can expect the response of a cell to be elaborate even when faced with a simple stress condition. A fundamental point in a network setting is that one should evaluate the role of individual players (such as a regulator) not in isolation, but with the knowledge that the entire network will react to what it does. In other words, local changes can have global effects. This, in turn, results in subtle cause–effect relationships. Studying the functions of a regulator by generating deletion or overexpression strains is often performed under the assumption that other regulators will remain static. In reality, however, such modifications can lead to aberrant changes across the network in ways that were initially unintended. Moreover, there is a possibility that such changes can occur because the network connections might not always be obvious. This can be attributed, in part, to as yet uneludicated implicit players in the network that dictate or otherwise influence cellular response. This is a likely explanation for the many ‘global’ regulators in P. aeruginosa and in similar bacteria that have complex regulatory networks. In such cases, many of the phenotypes observed with single regulator mutant strains can be part of a ripple effect that propagates through the network affecting disparate phenotypes.

A simplified model of gene regulatory network treats genes as being on or off, that is, taking binary values. It is, therefore, no surprise that Boolean networks (discrete dynamical network models) have been used to model and study gene regulatory circuits (319–321). Probabilistic Boolean networks, which take into account molecular and genetic noise (322,323), and stochastic Boolean networks, which permit the modelling of gene perturbations (324), provide important insights into the dynamical behaviour of the system. Although they are computationally complex, they are a valuable addition to the numerous other programs that are available to analyse gene regulatory networks (31,325,326).

Dynamical systems theory helps us to analyse the behaviour of complex systems that can frequently be expressed by time-differential equations. When the behaviour of a dynamical system depends sensitively on small changes in initial conditions, then the system is said to be chaotic, that is, capable of exhibiting chaotic behaviour. Researchers have investigated whether regulatory networks can have subsystems that are capable of exhibiting chaotic behaviour (327). It has been shown that competition between two or more subnetworks of comparable importance can lead to chaos (328–330). In fact, chaos has been shown to be possible in biochemical systems with only two feedback loops, and positive feedback is known to be necessary for chaotic behaviour (331). So, one would expect chaotic subsystems in a regulatory network as complex as the one that controls P. aeruginosa virulence (Figure 1). Despite of this predisposition, gene regulatory networks seldom exhibit chaotic behaviour. This could be because the competitions between opposing nodes are not strong enough (332) or that chaotic behaviours are short-lived because of triggering of other pathways, such as cell–cell communication (333). Another possibility is that the natural random variability of biochemical systems masks the chaotic behaviour (332). However, maintaining a low level of chaos in such a complex network is probably a combination of the aforementioned and, potentially, as yet unknown factors.

In gene regulatory networks, a particular dynamical system is characterized by time-evolving variables (chemical concentrations, gene expression and so forth) and by parameters (temperature, ambient chemical concentrations and so forth). A network can exhibit chaotic or non-chaotic behaviour depending on the parameters that influence it (334). Environmental factors, such as the temperature or the nutritional status of the cells, parameterize the relationship between transcription factors and the genes that they regulate. Although it is understood that some choices of parameters can induce chaotic behaviours, the parameter may not even be achievable, such as high temperatures (334). Mutations can also alter relationships in regulatory networks by causing changes in existing links or forming new ones. In a dynamically robust (non-chaotic) system, small finite changes in the parameters lead to only qualitative changes in the dynamical behaviour. However, there are boundaries in the parameter space where the behaviour of the system changes qualitatively and may include the possibility of chaotic dynamics. Predicting whether a network will be stable or chaos-prone under some conditions has been proven to be difficult and remains poorly characterized. Recent work has identified the minimum number, types and interactions among three and four nodes/subnetworks that can lead to chaos in a gene regulatory network (332). Such minimal subnetworks have been termed ‘chaotic motifs’, and networks with these motifs can exhibit chaotic behaviour under the right parameters (332). Analysis of the network in Figure 1 does not readily show such chaotic motifs. This could be because the network is incomplete (lack of data on the interactions among the P. aeruginosa regulators) or because of errors in the inferred interactions. Although P. aeruginosa virulence regulation has been extensively studied, there is yet much to learn. Thus, absence of empirical evidence does not preclude a propensity to chaos and is worth further investigation.

Depending on an elaborate network to achieve gene regulation is likely an adaptive mechanism by P. aeruginosa. Possessing alternate pathways to regulate the same phenotype ensures a rapid response to stimuli even if one of the pathways is affected, thus enhancing survival. Such examples can be seen throughout the network. As discussed in the ‘Toxins and Exoproteins’ section, expression of T3SS genes can be regulated at multiple levels, in response to various different signals and stress conditions, and it is not entirely dependent on any one signal. However, the extent of contributions of the individual regulators and, consequently, the fine balance that exists in some regulatory cascades within the network, are sometimes not easily apparent. Network dependence is also a probable reason for regulator genes being non-essential, in the sense that deleting a transcriptional regulator gene typically does not affect cell viability because of the presence of alternate regulatory mechanisms. Having key regulators modulate different related phenotypes has the added advantage in allowing the cells to adapt to external signals by modulating one or a few regulators instead of individually regulating different virulence systems. A case in point is AmpR that positively regulates acute virulence factors while downregulating chronic infection phenotypes (23). Also of importance is the co-regulation of metabolism and virulence. Studies have identified regulators like CbrB that, with its cognate sensor CbrA, not only regulate carbon metabolism but also virulence phenotypes through the rgRNA, rgCrcZ and the RNA-binding protein Crc (306,307). Moreover, there is crosstalk between CbrA and regulators other than CbrB, highlighting the complexity of the system (306).

The plethora of transcriptome studies using microarrays or deep sequencing will add to the database of genes that are differentially expressed in response to regulator mutations or specific growth conditions. Differentiating the direct effect of a change from a ripple effect can, at least partly, be achieved by meta-analysis studies that look at multiple transcriptomes, identifying effects unique to each condition and differentiating them from the so-called ripple (32,33). Network analyses can help us understand the relationship between different regulators, group them based on function and, more importantly, help identify critical nodes and prominent players. This can serve as a means of target identification in attempting to deal with P. aeruginosa infections. In Figure 1, we see that some parts of the network are more densely connected than others, with central cores containing most of the links. A case in point is LasR of the QS subnetwork. It is well known that QS is central to virulence regulation in P. aeruginosa and targeting key regulators will have a better chance of therapeutic success. Recently, inhibitors of a key QS regulator were shown to reduce pathogenicity in Vibrio cholera (335).

With the extensive use of high-throughput transcriptomics, gene regulation studies are now focusing on the role of non-coding RNAs in bacteria. rgRNAs have been shown to be extensively involved in gene regulation in P. aeruginosa and other bacteria (124,237,299,305,314,336). Techniques such as RNA-seq allow for the entire transcriptome to be sequenced, giving us an unprecedented insight into non-coding RNAs, asRNAs and sRNAs involved in regulation. Preliminary studies using prediction software and complementary experiments have already advanced our understanding (29,108,124,299,314,337). Given the many different ways in which small RNAs can modulate gene expression (313) and potentially undiscovered ones, we can look forward to exciting new discoveries in bacterial gene regulation in the coming years.

FUNDING

National Institutes of Health-Minority Medical Research Support SCORE [S06 GM08205 and 5SC1AI081376 to K.M.]; FIU Research Assistantship (Herbert Werthiem College of Medicine to D.B.). Funding for open access charge: National Institutes of Health-Minority Medical Research Support SCORE [5SC1AI081376 to K.M.].

Conflict of interest statement. None declared.

ACKNOWLEDGEMENTS

The authors are extremely grateful to Giri Narasimhan (Florida International University) and Edward Celarier (NASA Goddard) for extensive discussions and critical comments on chaos theory.

REFERENCES

1
Rahme
LG
Ausubel
FM
Cao
H
Drenkard
E
Goumnerov
BC
Lau
GW
Mahajan-Miklos
S
Plotnikova
J
Tan
MW
Tsongalis
J
, et al. 
Plants and animals share functionally common bacterial virulence factors
Proc. Natl Acad. Sci. USA
2000
, vol. 
97
 (pg. 
8815
-
8821
)
2
Mahajan-Miklos
S
Rahme
LG
Ausubel
FM
Elucidating the molecular mechanisms of bacterial virulence using non-mammalian hosts
Mol. Microbiol.
2000
, vol. 
37
 (pg. 
981
-
988
)
3
Kerr
KG
Snelling
AM
Pseudomonas aeruginosa: a formidable and ever-present adversary
J. Hosp. Infect.
2009
, vol. 
73
 (pg. 
338
-
344
)
4
Valderrey
AD
Pozuelo
MJ
Jimenez
PA
Macia
MD
Oliver
A
Rotger
R
Chronic colonization by Pseudomonas aeruginosa of patients with obstructive lung diseases: cystic fibrosis, bronchiectasis, and chronic obstructive pulmonary disease
Diagn. Microbiol. Infect. Dis.
2010
, vol. 
68
 (pg. 
20
-
27
)
5
Bouza
E
Burillo
A
Munoz
P
Catheter-related infections: diagnosis and intravascular treatment
Clin. Microbiol. Infect.
2002
, vol. 
8
 (pg. 
265
-
274
)
6
Manfredi
R
Nanetti
A
Ferri
M
Chiodo
F
Pseudomonas spp. complications in patients with HIV disease: an eight-year clinical and microbiological survey
Eur. J. Epidemiol.
2000
, vol. 
16
 (pg. 
111
-
118
)
7
Mizuno
T
Compilation of all genes encoding two-component phosphotransfer signal transducers in the genome of Escherichia coli
DNA Res.
1997
, vol. 
4
 (pg. 
161
-
168
)
8
Fabret
C
Feher
VA
Hoch
JA
Two-component signal transduction in Bacillus subtilis: how one organism sees its world
J. Bacteriol.
1999
, vol. 
181
 (pg. 
1975
-
1983
)
9
Gooderham
WJ
Hancock
RE
Regulation of virulence and antibiotic resistance by two-component regulatory systems in Pseudomonas aeruginosa
FEMS Microbiol. Rev.
2009
, vol. 
33
 (pg. 
279
-
294
)
10
Buelow
DR
Raivio
TL
Three (and more) component regulatory systems - auxiliary regulators of bacterial histidine kinases
Mol. Microbiol.
2010
, vol. 
75
 (pg. 
547
-
566
)
11
Ng
WL
Bassler
BL
Bacterial quorum-sensing network architectures
Annu. Rev. Genet.
2009
, vol. 
43
 (pg. 
197
-
222
)
12
Harmsen
M
Yang
L
Pamp
SJ
Tolker-Nielsen
T
An update on Pseudomonas aeruginosa biofilm formation, tolerance, and dispersal
FEMS Immunol. Med. Microbiol.
2010
, vol. 
59
 (pg. 
253
-
268
)
13
Njoroge
J
Sperandio
V
Jamming bacterial communication: new approaches for the treatment of infectious diseases
EMBO Mol. Med.
2009
, vol. 
1
 (pg. 
201
-
210
)
14
Burns
JL
Gibson
RL
McNamara
S
Yim
D
Emerson
J
Rosenfeld
M
Hiatt
P
McCoy
K
Castile
R
Smith
AL
, et al. 
Longitudinal assessment of Pseudomonas aeruginosa in young children with cystic fibrosis
J. Infect. Dis.
2001
, vol. 
183
 (pg. 
444
-
452
)
15
Hogardt
M
Heesemann
J
Microevolution of Pseudomonas aeruginosa to a chronic pathogen of the cystic fibrosis lung
Curr. Top. Microbiol. Immunol.
2012
 
Feb 8 (doi: 10.1007/82_2011_199; epub ahead of print)
16
Coggan
KA
Wolfgang
MC
Global regulatory pathways and cross-talk control Pseudomonas aeruginosa environmental lifestyle and virulence phenotype
Curr. Issues Mol. Biol.
2012
, vol. 
14
 (pg. 
47
-
70
)
17
Hoboth
C
Hoffmann
R
Eichner
A
Henke
C
Schmoldt
S
Imhof
A
Heesemann
J
Hogardt
M
Dynamics of adaptive microevolution of hypermutable Pseudomonas aeruginosa during chronic pulmonary infection in patients with cystic fibrosis
J. Infect. Dis.
2009
, vol. 
200
 (pg. 
118
-
130
)
18
Hogardt
M
Hoboth
C
Schmoldt
S
Henke
C
Bader
L
Heesemann
J
Stage-specific adaptation of hypermutable Pseudomonas aeruginosa isolates during chronic pulmonary infection in patients with cystic fibrosis
J. Infect. Dis.
2007
, vol. 
195
 (pg. 
70
-
80
)
19
Oliver
A
Mena
A
Bacterial hypermutation in cystic fibrosis, not only for antibiotic resistance
Clin. Microbiol. Infect.
2010
, vol. 
16
 (pg. 
798
-
808
)
20
Hassett
DJ
Sutton
MD
Schurr
MJ
Herr
AB
Caldwell
CC
Matu
JO
Pseudomonas aeruginosa hypoxic or anaerobic biofilm infections within cystic fibrosis airways
Trends Microbiol.
2009
, vol. 
17
 (pg. 
130
-
138
)
21
Goodman
AL
Kulasekara
B
Rietsch
A
Boyd
D
Smith
RS
Lory
S
A signaling network reciprocally regulates genes associated with acute infection and chronic persistence in Pseudomonas aeruginosa
Dev. Cell
2004
, vol. 
7
 (pg. 
745
-
754
)
22
Ventre
I
Goodman
AL
Vallet-Gely
I
Vasseur
P
Soscia
C
Molin
S
Bleves
S
Lazdunski
A
Lory
S
Filloux
A
Multiple sensors control reciprocal expression of Pseudomonas aeruginosa regulatory RNA and virulence genes
Proc. Natl Acad. Sci. USA
2006
, vol. 
103
 (pg. 
171
-
176
)
23
Balasubramanian
D
Schneper
L
Merighi
M
Smith
R
Narasimhan
G
Lory
S
Mathee
K
The regulatory repertoire of Pseudomonas aeruginosa AmpC ß-lactamase regulator AmpR includes virulence genes
PLoS One
2012
, vol. 
7
 pg. 
e34067
 
24
Stover
CK
Pham
XQ
Erwin
AL
Mizoguchi
SD
Warrener
P
Hickey
MJ
Brinkman
FS
Hufnagle
WO
Kowalik
DJ
Lagrou
M
, et al. 
Complete genome sequence of Pseudomonas aeruginosa PA01, an opportunistic pathogen
Nature
2000
, vol. 
406
 (pg. 
959
-
964
)
25
Mathee
K
Narasimhan
G
Valdes
C
Qiu
X
Matewish
JM
Koehrsen
M
Rokas
A
Yandava
CN
Engels
R
Zeng
E
, et al. 
Dynamics of Pseudomonas aeruginosa genome evolution
Proc. Natl Acad. Sci. USA
2008
, vol. 
105
 (pg. 
3100
-
3105
)
26
Winsor
GL
Lam
DK
Fleming
L
Lo
R
Whiteside
MD
Yu
NY
Hancock
RE
Brinkman
FS
Pseudomonas genome database: improved comparative analysis and population genomics capability for Pseudomonas genomes
Nucleic Acids Res.
2011
, vol. 
39
 (pg. 
D596
-
D600
)
27
Balasubramanian
D
Murugapiran
SK
Silva-Herzog
E
Schneper
L
Yang
X
Tatke
G
Narasimhan
G
Mathee
K
Babu
MM
Bacterial Gene Regulation and Transcriptional Networks
2013
United Kingdom
Caiser Academic Press
28
Potvin
E
Sanschagrin
F
Levesque
RC
Sigma factors in Pseudomonas aeruginosa
FEMS Microbiol. Rev.
2008
, vol. 
32
 (pg. 
38
-
55
)
29
Livny
J
Brencic
A
Lory
S
Waldor
MK
Identification of 17 Pseudomonas aeruginosa sRNAs and prediction of sRNA-encoding genes in 10 diverse pathogens using the bioinformatic tool sRNAPredict2
Nucleic Acids Res.
2006
, vol. 
34
 (pg. 
3484
-
3493
)
30
Galan-Vasquez
E
Luna
B
Martinez-Antonio
A
The regulatory network of Pseudomonas aeruginosa
Microb. Inform. Exp.
2011
, vol. 
1
 pg. 
3
 
31
Babu
MM
Teichmann
SA
Aravind
L
Evolutionary dynamics of prokaryotic transcriptional regulatory networks
J. Mol. Biol.
2006
, vol. 
358
 (pg. 
614
-
633
)
32
Balasubramanian
D
Mathee
K
Comparative transcriptome analyses of Pseudomonas aeruginosa
Hum. Genomics
2009
, vol. 
3
 (pg. 
349
-
361
)
33
Goodman
AL
Lory
S
Analysis of regulatory networks in Pseudomonas aeruginosa by genomewide transcriptional profiling
Curr. Opin. Microbiol.
2004
, vol. 
7
 (pg. 
39
-
44
)
34
Ong
CT
Corces
VG
Enhancer function: new insights into the regulation of tissue-specific gene expression
Nat. Rev. Genet.
2011
, vol. 
12
 (pg. 
283
-
293
)
35
Levine
M
Transcriptional enhancers in animal development and evolution
Curr. Biol.
2010
, vol. 
20
 (pg. 
R754
-
R763
)
36
Bulger
M
Groudine
M
Functional and mechanistic diversity of distal transcription enhancers
Cell
2011
, vol. 
144
 (pg. 
327
-
339
)
37
Ishihama
A
Prokaryotic genome regulation: multifactor promoters, multitarget regulators and hierarchic networks
FEMS Microbiol. Rev.
2010
, vol. 
34
 (pg. 
628
-
645
)
38
Storz
G
Vogel
J
Wassarman
KM
Regulation by small RNAs in bacteria: expanding frontiers
Mol. Cell
2011
, vol. 
43
 (pg. 
880
-
891
)
39
Waters
LS
Storz
G
Regulatory RNAs in bacteria
Cell
2009
, vol. 
136
 (pg. 
615
-
628
)
40
Reitzer
LJ
Magasanik
B
Transcription of glnA in E. coli is stimulated by activator bound to sites far from the promoter
Cell
1986
, vol. 
45
 (pg. 
785
-
792
)
41
Popham
DL
Szeto
D
Keener
J
Kustu
S
Function of a bacterial activator protein that binds to transcriptional enhancers
Science
1989
, vol. 
243
 (pg. 
629
-
635
)
42
Buck
M
Gallegos
MT
Studholme
DJ
Guo
Y
Gralla
JD
The bacterial enhancer-dependent sigma(54) (sigma(N)) transcription factor
J. Bacteriol.
2000
, vol. 
182
 (pg. 
4129
-
4136
)
43
Mohr
CD
Leveau
JH
Krieg
DP
Hibler
NS
Deretic
V
AlgR-binding sites within the algD promoter make up a set of inverted repeats separated by a large intervening segment of DNA
J. Bacteriol.
1992
, vol. 
174
 (pg. 
6624
-
6633
)
44
Ramsey
DM
Baynham
PJ
Wozniak
DJ
Binding of Pseudomonas aeruginosa AlgZ to sites upstream of the algZ promoter leads to repression of transcription
J. Bacteriol.
2005
, vol. 
187
 (pg. 
4430
-
4443
)
45
Winteler
HV
Haas
D
The homologous regulators ANR of Pseudomonas aeruginosa and FNR of Escherichia coli have overlapping but distinct specificities for anaerobically inducible promoters
Microbiology
1996
, vol. 
142
 (pg. 
685
-
693
)
46
Lu
CD
Winteler
H
Abdelal
A
Haas
D
The ArgR regulatory protein, a helper to the anaerobic regulator ANR during transcriptional activation of the arcD promoter in Pseudomonas aeruginosa
J. Bacteriol.
1999
, vol. 
181
 (pg. 
2459
-
2464
)
47
Hovey
AK
Frank
DW
Analyses of the DNA-binding and transcriptional activation properties of ExsA, the transcriptional activator of the Pseudomonas aeruginosa exoenzyme S regulon
J. Bacteriol.
1995
, vol. 
177
 (pg. 
4427
-
4436
)
48
Baraquet
C
Murakami
K
Parsek
MR
Harwood
CS
The FleQ protein from Pseudomonas aeruginosa functions as both a repressor and an activator to control gene expression from the pel operon promoter in response to c-di-GMP
Nucleic Acids Res.
2012
, vol. 
40
 (pg. 
7207
-
7218
)
49
Ochsner
UA
Vasil
AI
Vasil
ML
Role of the ferric uptake regulator of Pseudomonas aeruginosa in the regulation of siderophores and exotoxin A expression: purification and activity on iron-regulated promoters
J. Bacteriol.
1995
, vol. 
177
 (pg. 
7194
-
7201
)
50
Wagner
VE
Bushnell
D
Passador
L
Brooks
AI
Iglewski
BH
Microarray analysis of Pseudomonas aeruginosa quorum-sensing regulons: effects of growth phase and environment
J. Bacteriol.
2003
, vol. 
185
 (pg. 
2080
-
2095
)
51
Tian
ZX
Fargier
E
Mac Aogain
M
Adams
C
Wang
YP
O'Gara
F
Transcriptome profiling defines a novel regulon modulated by the LysR-type transcriptional regulator MexT in Pseudomonas aeruginosa
Nucleic Acids Res.
2009
, vol. 
37
 (pg. 
7546
-
7559
)
52
Xiao
G
He
J
Rahme
LG
Mutation analysis of the Pseudomonas aeruginosa mvfR and pqsABCDE gene promoters demonstrates complex quorum-sensing circuitry
Microbiology
2006
, vol. 
152
 (pg. 
1679
-
1686
)
53
Kojic
M
Aguilar
C
Venturi
V
TetR family member psrA directly binds the Pseudomonas rpoS and psrA promoters
J. Bacteriol.
2002
, vol. 
184
 (pg. 
2324
-
2330
)
54
Nicastro
GG
Boechat
AL
Abe
CM
Kaihami
GH
Baldini
RL
Pseudomonas aeruginosa PA14 cupD transcription is activated by the RcsB response regulator, but repressed by its putative cognate sensor RcsC
FEMS Microbiol. Lett.
2009
, vol. 
301
 (pg. 
115
-
123
)
55
Pearson
JP
Pesci
EC
Iglewski
BH
Roles of Pseudomonas aeruginosa las and rhl quorum-sensing systems in control of elastase and rhamnolipid biosynthesis genes
J. Bacteriol.
1997
, vol. 
179
 (pg. 
5756
-
5767
)
56
Dasgupta
N
Ferrell
EP
Kanack
KJ
West
SE
Ramphal
R
fleQ, the gene encoding the major flagellar regulator of Pseudomonas aeruginosa, is sigma70 dependent and is downregulated by Vfr, a homolog of Escherichia coli cyclic AMP receptor protein
J. Bacteriol.
2002
, vol. 
184
 (pg. 
5240
-
5250
)
57
Liang
H
Deng
X
Ji
Q
Sun
F
Shen
T
He
C
The Pseudomonas aeruginosa global regulator VqsR directly inhibits QscR to control quorum-sensing and virulence gene expression
J. Bacteriol.
2012
, vol. 
194
 (pg. 
3098
-
3108
)
58
Nealson
KH
Platt
T
Hastings
JW
Cellular control of the synthesis and activity of the bacterial luminescent system
J. Bacteriol.
1970
, vol. 
104
 (pg. 
313
-
322
)
59
Stevens
AM
Schuster
M
Rumbaugh
KP
Working together for the common good: cell-cell communication in bacteria
J. Bacteriol.
2012
, vol. 
194
 (pg. 
2131
-
2141
)
60
Williams
P
Camara
M
Quorum sensing and environmental adaptation in Pseudomonas aeruginosa: a tale of regulatory networks and multifunctional signal molecules
Curr. Opin. Microbiol.
2009
, vol. 
12
 (pg. 
182
-
191
)
61
Jimenez
PN
Koch
G
Thompson
JA
Xavier
KB
Cool
RH
Quax
WJ
The multiple signaling systems regulating virulence in Pseudomonas aeruginosa
Microbiol. Mol. Biol. Rev.
2012
, vol. 
76
 (pg. 
46
-
65
)
62
Schuster
M
Greenberg
EP
A network of networks: quorum-sensing gene regulation in Pseudomonas aeruginosa
Int. J. Med. Microbiol.
2006
, vol. 
296
 (pg. 
73
-
81
)
63
Passador
L
Cook
JM
Gambello
MJ
Rust
L
Iglewski
BH
Expression of Pseudomonas aeruginosa virulence genes requires cell-to-cell communication
Science
1993
, vol. 
260
 (pg. 
1127
-
1130
)
64
Pearson
JP
Gray
KM
Passador
L
Tucker
KD
Eberhard
A
Iglewski
BH
Greenberg
EP
Structure of the autoinducer required for expression of Pseudomonas aeruginosa virulence genes
Proc. Natl Acad. Sci. USA
1994
, vol. 
91
 (pg. 
197
-
201
)
65
Brint
JM
Ohman
DE
Synthesis of multiple exoproducts in Pseudomonas aeruginosa is under the control of RhlR-RhlI, another set of regulators in strain PAO1 with homology to the autoinducer-responsive LuxR-LuxI family
J. Bacteriol.
1995
, vol. 
177
 (pg. 
7155
-
7163
)
66
Latifi
A
Winson
MK
Foglino
M
Bycroft
BW
Stewart
GS
Lazdunski
A
Williams
P
Multiple homologues of LuxR and LuxI control expression of virulence determinants and secondary metabolites through quorum sensing in Pseudomonas aeruginosa PAO1
Mol. Microbiol.
1995
, vol. 
17
 (pg. 
333
-
343
)
67
Ochsner
UA
Reiser
J
Autoinducer-mediated regulation of rhamnolipid biosurfactant synthesis in Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
1995
, vol. 
92
 (pg. 
6424
-
6428
)
68
Pearson
JP
Passador
L
Iglewski
BH
Greenberg
EP
A second N-acylhomoserine lactone signal produced by Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
1995
, vol. 
92
 (pg. 
1490
-
1494
)
69
Kiratisin
P
Tucker
KD
Passador
L
LasR, a transcriptional activator of Pseudomonas aeruginosa virulence genes, functions as a multimer
J. Bacteriol.
2002
, vol. 
184
 (pg. 
4912
-
4919
)
70
Lamb
JR
Patel
H
Montminy
T
Wagner
VE
Iglewski
BH
Functional domains of the RhlR transcriptional regulator of Pseudomonas aeruginosa
J. Bacteriol.
2003
, vol. 
185
 (pg. 
7129
-
7139
)
71
Schuster
M
Urbanowski
ML
Greenberg
EP
Promoter specificity in Pseudomonas aeruginosa quorum sensing revealed by DNA binding of purified LasR
Proc. Natl Acad. Sci. USA
2004
, vol. 
101
 (pg. 
15833
-
15839
)
72
Schuster
M
Greenberg
EP
Early activation of quorum sensing in Pseudomonas aeruginosa reveals the architecture of a complex regulon
BMC Genomics
2007
, vol. 
8
 pg. 
287
 
73
Diggle
SP
Matthijs
S
Wright
VJ
Fletcher
MP
Chhabra
SR
Lamont
IL
Kong
X
Hider
RC
Cornelis
P
Camara
M
, et al. 
The Pseudomonas aeruginosa 4-quinolone signal molecules HHQ and PQS play multifunctional roles in quorum sensing and iron entrapment
Chem. Biol.
2007
, vol. 
14
 (pg. 
87
-
96
)
74
Xiao
G
Deziel
E
He
J
Lepine
F
Lesic
B
Castonguay
MH
Milot
S
Tampakaki
AP
Stachel
SE
Rahme
LG
MvfR, a key Pseudomonas aeruginosa pathogenicity LTTR-class regulatory protein, has dual ligands
Mol. Microbiol.
2006
, vol. 
62
 (pg. 
1689
-
1699
)
75
Deziel
E
Gopalan
S
Tampakaki
AP
Lepine
F
Padfield
KE
Saucier
M
Xiao
G
Rahme
LG
The contribution of MvfR to Pseudomonas aeruginosa pathogenesis and quorum sensing circuitry regulation: multiple quorum sensing-regulated genes are modulated without affecting lasRI, rhlRI or the production of N-acyl-L-homoserine lactones
Mol. Microbiol.
2005
, vol. 
55
 (pg. 
998
-
1014
)
76
Bredenbruch
F
Geffers
R
Nimtz
M
Buer
J
Haussler
S
The Pseudomonas aeruginosa quinolone signal (PQS) has an iron-chelating activity
Environ. Microbiol.
2006
, vol. 
8
 (pg. 
1318
-
1329
)
77
Mashburn
LM
Whiteley
M
Membrane vesicles traffic signals and facilitate group activities in a prokaryote
Nature
2005
, vol. 
437
 (pg. 
422
-
425
)
78
Schertzer
JW
Whiteley
M
A bilayer-couple model of bacterial outer membrane vesicle biogenesis
MBio
2012
, vol. 
3
 (pg. 
e00297
-
11
)
79
Liang
H
Li
L
Dong
Z
Surette
MG
Duan
K
The YebC family protein PA0964 negatively regulates the Pseudomonas aeruginosa quinolone signal system and pyocyanin production
J. Bacteriol.
2008
, vol. 
190
 (pg. 
6217
-
6227
)
80
Wagner
VE
Li
LL
Isabella
VM
Iglewski
BH
Analysis of the hierarchy of quorum-sensing regulation in Pseudomonas aeruginosa
Anal. Bioanal. Chem.
2007
, vol. 
387
 (pg. 
469
-
479
)
81
Latifi
A
Foglino
M
Tanaka
K
Williams
P
Lazdunski
A
A hierarchical quorum-sensing cascade in Pseudomonas aeruginosa links the transcriptional activators LasR and RhIR (VsmR) to expression of the stationary-phase sigma factor RpoS
Mol. Microbiol.
1996
, vol. 
21
 (pg. 
1137
-
1146
)
82
McGrath
S
Wade
DS
Pesci
EC
Dueling quorum sensing systems in Pseudomonas aeruginosa control the production of the Pseudomonas quinolone signal (PQS)
FEMS Microbiol. Lett.
2004
, vol. 
230
 (pg. 
27
-
34
)
83
Pesci
EC
Pearson
JP
Seed
PC
Iglewski
BH
Regulation of las and rhl quorum sensing in Pseudomonas aeruginosa
J. Bacteriol.
1997
, vol. 
179
 (pg. 
3127
-
3132
)
84
Dekimpe
V
Deziel
E
Revisiting the quorum-sensing hierarchy in Pseudomonas aeruginosa: the transcriptional regulator RhlR regulates LasR-specific factors
Microbiology
2009
, vol. 
155
 (pg. 
712
-
723
)
85
Duan
K
Surette
MG
Environmental regulation of Pseudomonas aeruginosa PAO1 Las and Rhl quorum-sensing systems
J. Bacteriol.
2007
, vol. 
189
 (pg. 
4827
-
4836
)
86
Schuster
M
Hawkins
AC
Harwood
CS
Greenberg
EP
The Pseudomonas aeruginosa RpoS regulon and its relationship to quorum sensing
Mol. Microbiol.
2004
, vol. 
51
 (pg. 
973
-
985
)
87
Gilbert
KB
Kim
TH
Gupta
R
Greenberg
EP
Schuster
M
Global position analysis of the Pseudomonas aeruginosa quorum-sensing transcription factor LasR
Mol. Microbiol.
2009
, vol. 
73
 (pg. 
1072
-
1085
)
88
Castang
S
McManus
HR
Turner
KH
Dove
SL
H-NS family members function coordinately in an opportunistic pathogen
Proc. Natl Acad. Sci. USA
2008
, vol. 
105
 (pg. 
18947
-
18952
)
89
Rampioni
G
Schuster
M
Greenberg
EP
Bertani
I
Grasso
M
Venturi
V
Zennaro
E
Leoni
L
RsaL provides quorum sensing homeostasis and functions as a global regulator of gene expression in Pseudomonas aeruginosa
Mol. Microbiol.
2007
, vol. 
66
 (pg. 
1557
-
1565
)
90
Rampioni
G
Schuster
M
Greenberg
EP
Zennaro
E
Leoni
L
Contribution of the RsaL global regulator to Pseudomonas aeruginosa virulence and biofilm formation
FEMS Microbiol. Lett.
2009
, vol. 
301
 (pg. 
210
-
217
)
91
Wei
Q
Le Minh
PN
Dotsch
A
Hildebrand
F
Panmanee
W
Elfarash
A
Schulz
S
Plaisance
S
Charlier
D
Hassett
D
, et al. 
Global regulation of gene expression by OxyR in an important human opportunistic pathogen
Nucleic Acids Res.
2012
, vol. 
40
 (pg. 
4320
-
4333
)
92
Dieppois
G
Ducret
V
Caille
O
Perron
K
The transcriptional regulator CzcR modulates antibiotic resistance and quorum sensing in Pseudomonas aeruginosa
PLoS One
2012
, vol. 
7
 pg. 
e38148
 
93
Perron
K
Caille
O
Rossier
C
Van Delden
C
Dumas
JL
Kohler
T
CzcR-CzcS, a two-component system involved in heavy metal and carbapenem resistance in Pseudomonas aeruginosa
J. Biol. Chem.
2004
, vol. 
279
 (pg. 
8761
-
8768
)
94
Juhas
M
Wiehlmann
L
Huber
B
Jordan
D
Lauber
J
Salunkhe
P
Limpert
AS
von Gotz
F
Steinmetz
I
Eberl
L
, et al. 
Global regulation of quorum sensing and virulence by VqsR in Pseudomonas aeruginosa
Microbiology
2004
, vol. 
150
 (pg. 
831
-
841
)
95
Li
LL
Malone
JE
Iglewski
BH
Regulation of the Pseudomonas aeruginosa quorum-sensing regulator VqsR
J. Bacteriol.
2007
, vol. 
189
 (pg. 
4367
-
4374
)
96
Liang
H
Deng
X
Ji
Q
Sun
F
Shen
T
He
C
The Pseudomonas aeruginosa global regulator VqsR directly inhibits QscR to control quorum-sensing and virulence gene expression
J. Bacteriol.
2012
, vol. 
194
 (pg. 
3098
-
3108
)
97
Lee
JH
Lequette
Y
Greenberg
EP
Activity of purified QscR, a Pseudomonas aeruginosa orphan quorum-sensing transcription factor
Mol. Microbiol.
2006
, vol. 
59
 (pg. 
602
-
609
)
98
Lequette
Y
Lee
JH
Ledgham
F
Lazdunski
A
Greenberg
EP
A distinct QscR regulon in the Pseudomonas aeruginosa quorum-sensing circuit
J. Bacteriol.
2006
, vol. 
188
 (pg. 
3365
-
3370
)
99
Ledgham
F
Ventre
I
Soscia
C
Foglino
M
Sturgis
JN
Lazdunski
A
Interactions of the quorum sensing regulator QscR: interaction with itself and the other regulators of Pseudomonas aeruginosa LasR and RhlR
Mol. Microbiol.
2003
, vol. 
48
 (pg. 
199
-
210
)
100
Chugani
SA
Whiteley
M
Lee
KM
D'Argenio
D
Manoil
C
Greenberg
EP
QscR, a modulator of quorum-sensing signal synthesis and virulence in Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
2001
, vol. 
98
 (pg. 
2752
-
2757
)
101
Dong
YH
Zhang
XF
Xu
JL
Tan
AT
Zhang
LH
VqsM, a novel AraC-type global regulator of quorum-sensing signalling and virulence in Pseudomonas aeruginosa
Mol. Microbiol.
2005
, vol. 
58
 (pg. 
552
-
564
)
102
Deziel
E
Lepine
F
Milot
S
He
J
Mindrinos
MN
Tompkins
RG
Rahme
LG
Analysis of Pseudomonas aeruginosa 4-hydroxy-2-alkylquinolines (HAQs) reveals a role for 4-hydroxy-2-heptylquinoline in cell-to-cell communication
Proc. Natl Acad. Sci. USA
2004
, vol. 
101
 (pg. 
1339
-
1344
)
103
Gallagher
LA
McKnight
SL
Kuznetsova
MS
Pesci
EC
Manoil
C
Functions required for extracellular quinolone signaling by Pseudomonas aeruginosa
J. Bacteriol.
2002
, vol. 
184
 (pg. 
6472
-
6480
)
104
Farrow
JM
3rd
Pesci
EC
Two distinct pathways supply anthranilate as a precursor of the Pseudomonas quinolone signal
J. Bacteriol.
2007
, vol. 
189
 (pg. 
3425
-
3433
)
105
Chugani
S
Greenberg
EP
LuxR homolog-independent gene regulation by acyl-homoserine lactones in Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
2010
, vol. 
107
 (pg. 
10673
-
10678
)
106
Siehnel
R
Traxler
B
An
DD
Parsek
MR
Schaefer
AL
Singh
PK
A unique regulator controls the activation threshold of quorum-regulated genes in Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
2010
, vol. 
107
 (pg. 
7916
-
7921
)
107
Pessi
G
Williams
F
Hindle
Z
Heurlier
K
Holden
MT
Camara
M
Haas
D
Williams
P
The global posttranscriptional regulator RsmA modulates production of virulence determinants and N-acylhomoserine lactones in Pseudomonas aeruginosa
J. Bacteriol.
2001
, vol. 
183
 (pg. 
6676
-
6683
)
108
Sonnleitner
E
Schuster
M
Sorger-Domenigg
T
Greenberg
EP
Blasi
U
Hfq-dependent alterations of the transcriptome profile and effects on quorum sensing in Pseudomonas aeruginosa
Mol. Microbiol.
2006
, vol. 
59
 (pg. 
1542
-
1558
)
109
Kong
KF
Jayawardena
SR
Indulkar
SD
Del Puerto
A
Koh
CL
Hoiby
N
Mathee
K
Pseudomonas aeruginosa AmpR is a global transcriptional factor that regulates expression of AmpC and PoxB beta-lactamases, proteases, quorum sensing, and other virulence factors
Antimicrob. Agents Chemother.
2005
, vol. 
49
 (pg. 
4567
-
4575
)
110
Balasubramanian
D
Kong
KF
Jayawardena
SR
Leal
SM
Sautter
RT
Mathee
K
Co-regulation of ß-lactam resistance, alginate production and quorum sensing in Pseudomonas aeruginosa
J. Med. Microbiol.
2011
, vol. 
60
 (pg. 
147
-
156
)
111
Rodrigue
A
Quentin
Y
Lazdunski
A
Mejean
V
Foglino
M
Two-component systems in Pseudomonas aeruginosa: why so many?
Trends Microbiol.
2000
, vol. 
8
 (pg. 
498
-
504
)
112
Raghavan
V
Groisman
EA
Orphan and hybrid two-component system proteins in health and disease
Curr. Opin. Microbiol.
2010
, vol. 
13
 (pg. 
226
-
231
)
113
Stock
AM
Robinson
VL
Goudreau
PN
Two-component signal transduction
Annu. Rev. Biochem.
2000
, vol. 
69
 (pg. 
183
-
215
)
114
Laub
MT
Goulian
M
Specificity in two-component signal transduction pathways
Annu. Rev. Genet.
2007
, vol. 
41
 (pg. 
121
-
145
)
115
Mascher
T
Helmann
JD
Unden
G
Stimulus perception in bacterial signal-transducing histidine kinases
Microbiol. Mol. Biol. Rev.
2006
, vol. 
70
 (pg. 
910
-
938
)
116
Gao
R
Stock
AM
Molecular strategies for phosphorylation-mediated regulation of response regulator activity
Curr. Opin. Microbiol.
2010
, vol. 
13
 (pg. 
160
-
167
)
117
Mitrophanov
AY
Groisman
EA
Signal integration in bacterial two-component regulatory systems
Genes Dev.
2008
, vol. 
22
 (pg. 
2601
-
2611
)
118
Kitten
T
Kinscherf
TG
McEvoy
JL
Willis
DK
A newly identified regulator is required for virulence and toxin production in Pseudomonas syringae
Mol. Microbiol.
1998
, vol. 
28
 (pg. 
917
-
929
)
119
Coleman
FT
Mueschenborn
S
Meluleni
G
Ray
C
Carey
VJ
Vargas
SO
Cannon
CL
Ausubel
FM
Pier
GB
Hypersusceptibility of cystic fibrosis mice to chronic Pseudomonas aeruginosa oropharyngeal colonization and lung infection
Proc. Natl Acad. Sci. USA
2003
, vol. 
100
 (pg. 
1949
-
1954
)
120
Hrabak
EM
Willis
DK
The lemA gene required for pathogenicity of Pseudomonas syringae pv. syringae on bean is a member of a family of two-component regulators
J. Bacteriol.
1992
, vol. 
174
 (pg. 
3011
-
3020
)
121
Goodman
AL
Merighi
M
Hyodo
M
Ventre
I
Filloux
A
Lory
S
Direct interaction between sensor kinase proteins mediates acute and chronic disease phenotypes in a bacterial pathogen
Genes Dev.
2009
, vol. 
23
 (pg. 
249
-
259
)
122
Heeb
S
Haas
D
Regulatory roles of the GacS/GacA two-component system in plant-associated and other Gram-negative bacteria
Mol. Plant Microbe Interact.
2001
, vol. 
14
 (pg. 
1351
-
1363
)
123
Wolfgang
MC
Lee
VT
Gilmore
ME
Lory
S
Coordinate regulation of bacterial virulence genes by a novel adenylate cyclase-dependent signaling pathway
Dev. Cell
2003
, vol. 
4
 (pg. 
253
-
263
)
124
Brencic
A
Lory
S
Determination of the regulon and identification of novel mRNA targets of Pseudomonas aeruginosa RsmA
Mol. Microbiol.
2009
, vol. 
72
 (pg. 
612
-
632
)
125
Linares
JF
Gustafsson
I
Baquero
F
Martinez
JL
Antibiotics as intermicrobial signaling agents instead of weapons
Proc. Natl Acad. Sci. USA
2006
, vol. 
103
 (pg. 
19484
-
19489
)
126
McPhee
JB
Bains
M
Winsor
G
Lewenza
S
Kwasnicka
A
Brazas
MD
Brinkman
FS
Hancock
RE
Contribution of the PhoP-PhoQ and PmrA-PmrB two-component regulatory systems to Mg2+-induced gene regulation in Pseudomonas aeruginosa
J. Bacteriol.
2006
, vol. 
188
 (pg. 
3995
-
4006
)
127
Macfarlane
EL
Kwasnicka
A
Hancock
RE
Role of Pseudomonas aeruginosa PhoP-PhoQ in resistance to antimicrobial cationic peptides and aminoglycosides
Microbiology
2000
, vol. 
146
 (pg. 
2543
-
2554
)
128
Gooderham
WJ
Gellatly
SL
Sanschagrin
F
McPhee
JB
Bains
M
Cosseau
C
Levesque
RC
Hancock
RE
The sensor kinase PhoQ mediates virulence in Pseudomonas aeruginosa
Microbiology
2009
, vol. 
155
 (pg. 
699
-
711
)
129
McPhee
JB
Lewenza
S
Hancock
RE
Cationic antimicrobial peptides activate a two-component regulatory system, PmrA-PmrB, that regulates resistance to polymyxin B and cationic antimicrobial peptides in Pseudomonas aeruginosa
Mol. Microbiol.
2003
, vol. 
50
 (pg. 
205
-
217
)
130
Kwon
DH
Lu
CD
Polyamines induce resistance to cationic peptide, aminoglycoside, and quinolone antibiotics in Pseudomonas aeruginosa PAO1
Antimicrob. Agents Chemother.
2006
, vol. 
50
 (pg. 
1615
-
1622
)
131
Kwon
DH
Lu
CD
Polyamine effects on antibiotic susceptibility in bacteria
Antimicrob. Agents Chemother.
2007
, vol. 
51
 (pg. 
2070
-
2077
)
132
Flemming
HC
Wingender
J
The biofilm matrix
Nat. Rev. Microbiol.
2010
, vol. 
8
 (pg. 
623
-
633
)
133
Donlan
RM
Costerton
JW
Biofilms: survival mechanisms of clinically relevant microorganisms
Clin. Microbiol. Rev.
2002
, vol. 
15
 (pg. 
167
-
193
)
134
Costerton
JW
Stewart
PS
Greenberg
EP
Bacterial biofilms: a common cause of persistent infections
Science
1999
, vol. 
284
 (pg. 
1318
-
1322
)
135
Lopez
D
Vlamakis
H
Kolter
R
Biofilms
Cold Spring Harb Perspect. Biol.
2010
, vol. 
2
 pg. 
a000398
 
136
Stapper
AP
Narasimhan
G
Ohman
DE
Barakat
J
Hentzer
M
Molin
S
Kharazmi
A
Hoiby
N
Mathee
K
Alginate production affects Pseudomonas aeruginosa biofilm development and architecture, but is not essential for biofilm formation
J. Med. Microbiol.
2004
, vol. 
53
 (pg. 
679
-
690
)
137
Klausen
M
Heydorn
A
Ragas
P
Lambertsen
L
Aaes-Jorgensen
A
Molin
S
Tolker-Nielsen
T
Biofilm formation by Pseudomonas aeruginosa wild type, flagella and type IV pili mutants
Mol. Microbiol.
2003
, vol. 
48
 (pg. 
1511
-
1524
)
138
Caiazza
NC
O'Toole
GA
SadB is required for the transition from reversible to irreversible attachment during biofilm formation by Pseudomonas aeruginosa PA14
J. Bacteriol.
2004
, vol. 
186
 (pg. 
4476
-
4485
)
139
Merritt
JH
Brothers
KM
Kuchma
SL
O'Toole
GA
SadC reciprocally influences biofilm formation and swarming motility via modulation of exopolysaccharide production and flagellar function
J. Bacteriol.
2007
, vol. 
189
 (pg. 
8154
-
8164
)
140
Vallet
I
Olson
JW
Lory
S
Lazdunski
A
Filloux
A
The chaperone/usher pathways of Pseudomonas aeruginosa: identification of fimbrial gene clusters (cup) and their involvement in biofilm formation
Proc. Natl Acad. Sci. USA
2001
, vol. 
98
 (pg. 
6911
-
6916
)
141
Vallet
I
Diggle
SP
Stacey
RE
Camara
M
Ventre
I
Lory
S
Lazdunski
A
Williams
P
Filloux
A
Biofilm formation in Pseudomonas aeruginosa: fimbrial cup gene clusters are controlled by the transcriptional regulator MvaT
J. Bacteriol.
2004
, vol. 
186
 (pg. 
2880
-
2890
)
142
Vallet-Gely
I
Donovan
KE
Fang
R
Joung
JK
Dove
SL
Repression of phase-variable cup gene expression by H-NS-like proteins in Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
2005
, vol. 
102
 (pg. 
11082
-
11087
)
143
Kulasekara
HD
Ventre
I
Kulasekara
BR
Lazdunski
A
Filloux
A
Lory
S
A novel two-component system controls the expression of Pseudomonas aeruginosa fimbrial cup genes
Mol. Microbiol.
2005
, vol. 
55
 (pg. 
368
-
380
)
144
Merkel
TJ
Barros
C
Stibitz
S
Characterization of the bvgR locus of Bordetella pertussis
J. Bacteriol.
1998
, vol. 
180
 (pg. 
1682
-
1690
)
145
Kuchma
SL
Connolly
JP
O'Toole
GA
A three-component regulatory system regulates biofilm maturation and type III secretion in Pseudomonas aeruginosa
J. Bacteriol.
2005
, vol. 
187
 (pg. 
1441
-
1454
)
146
Mikkelsen
H
Ball
G
Giraud
C
Filloux
A
Expression of Pseudomonas aeruginosa CupD fimbrial genes is antagonistically controlled by RcsB and the EAL-containing PvrR response regulators
PLoS One
2009
, vol. 
4
 pg. 
e6018
 
147
Hickman
JW
Tifrea
DF
Harwood
CS
A chemosensory system that regulates biofilm formation through modulation of cyclic diguanylate levels
Proc. Natl Acad. Sci. USA
2005
, vol. 
102
 (pg. 
14422
-
14427
)
148
Guvener
ZT
Harwood
CS
Subcellular location characteristics of the Pseudomonas aeruginosa GGDEF protein, WspR, indicate that it produces cyclic-di-GMP in response to growth on surfaces
Mol. Microbiol.
2007
, vol. 
66
 (pg. 
1459
-
1473
)
149
Choy
WK
Zhou
L
Syn
CK
Zhang
LH
Swarup
S
MorA defines a new class of regulators affecting flagellar development and biofilm formation in diverse Pseudomonas species
J. Bacteriol.
2004
, vol. 
186
 (pg. 
7221
-
7228
)
150
Ueda
A
Wood
TK
Connecting quorum sensing, c-di-GMP, pel polysaccharide, and biofilm formation in Pseudomonas aeruginosa through tyrosine phosphatase TpbA (PA3885)
PLoS Pathog.
2009
, vol. 
5
 pg. 
e1000483
 
151
Hickman
JW
Harwood
CS
Identification of FleQ from Pseudomonas aeruginosa as a c-di-GMP-responsive transcription factor
Mol. Microbiol.
2008
, vol. 
69
 (pg. 
376
-
389
)
152
Attila
C
Ueda
A
Wood
TK
PA2663 (PpyR) increases biofilm formation in Pseudomonas aeruginosa PAO1 through the psl operon and stimulates virulence and quorum-sensing phenotypes
Appl. Microbiol. Biotechnol.
2008
, vol. 
78
 (pg. 
293
-
307
)
153
D'Argenio
DA
Calfee
MW
Rainey
PB
Pesci
EC
Autolysis and autoaggregation in Pseudomonas aeruginosa colony morphology mutants
J. Bacteriol.
2002
, vol. 
184
 (pg. 
6481
-
6489
)
154
Allesen-Holm
M
Barken
KB
Yang
L
Klausen
M
Webb
JS
Kjelleberg
S
Molin
S
Givskov
M
Tolker-Nielsen
T
A characterization of DNA release in Pseudomonas aeruginosa cultures and biofilms
Mol. Microbiol.
2006
, vol. 
59
 (pg. 
1114
-
1128
)
155
Heurlier
K
Denervaud
V
Haenni
M
Guy
L
Krishnapillai
V
Haas
D
Quorum-sensing-negative (lasR) mutants of Pseudomonas aeruginosa avoid cell lysis and death
J. Bacteriol.
2005
, vol. 
187
 (pg. 
4875
-
4883
)
156
Whitchurch
CB
Tolker-Nielsen
T
Ragas
PC
Mattick
JS
Extracellular DNA required for bacterial biofilm formation
Science
2002
, vol. 
295
 pg. 
1487
 
157
Dominiak
DM
Nielsen
JL
Nielsen
PH
Extracellular DNA is abundant and important for microcolony strength in mixed microbial biofilms
Environ. Microbiol.
2010
, vol. 
13
 (pg. 
710
-
721
)
158
Pamp
SJ
Tolker-Nielsen
T
Multiple roles of biosurfactants in structural biofilm development by Pseudomonas aeruginosa
J. Bacteriol.
2007
, vol. 
189
 (pg. 
2531
-
2539
)
159
Davey
ME
Caiazza
NC
O'Toole
GA
Rhamnolipid surfactant production affects biofilm architecture in Pseudomonas aeruginosa PAO1
J. Bacteriol.
2003
, vol. 
185
 (pg. 
1027
-
1036
)
160
Petrova
OE
Sauer
K
A novel signaling network essential for regulating Pseudomonas aeruginosa biofilm development
PLoS Pathog.
2009
, vol. 
5
 pg. 
e1000668
 
161
Petrova
OE
Sauer
K
SagS contributes to the motile-sessile switch and acts in concert with BfiSR to enable Pseudomonas aeruginosa biofilm formation
J. Bacteriol.
2011
, vol. 
193
 (pg. 
6614
-
6628
)
162
Janjua
HA
Segata
N
Bernabo
P
Tamburini
S
Ellen
A
Jousson
O
Clinical populations of Pseudomonas aeruginosa isolated from acute infections show a wide virulence range partially correlated with population structure and virulence gene expression
Microbiology
2012
, vol. 
158
 (pg. 
2089
-
2098
)
163
Storey
DG
Ujack
EE
Rabin
HR
Mitchell
I
Pseudomonas aeruginosa lasR transcription correlates with the transcription of lasA, lasB, and toxA in chronic lung infections associated with cystic fibrosis
Infect. Immun.
1998
, vol. 
66
 (pg. 
2521
-
2528
)
164
Singh
PK
Schaefer
AL
Parsek
MR
Moninger
TO
Welsh
MJ
Greenberg
EP
Quorum-sensing signals indicate that cystic fibrosis lungs are infected with bacterial biofilms
Nature
2000
, vol. 
407
 (pg. 
762
-
764
)
165
Wu
H
Song
Z
Givskov
M
Doring
G
Worlitzsch
D
Mathee
K
Rygaard
J
Hoiby
N
Pseudomonas aeruginosa mutations in lasI and rhlI quorum sensing systems result in milder chronic lung infection
Microbiology
2001
, vol. 
147
 (pg. 
1105
-
1113
)
166
Bjarnsholt
T
Jensen
PO
Burmolle
M
Hentzer
M
Haagensen
JA
Hougen
HP
Calum
H
Madsen
KG
Moser
C
Molin
S
, et al. 
Pseudomonas aeruginosa tolerance to tobramycin, hydrogen peroxide and polymorphonuclear leukocytes is quorum-sensing dependent
Microbiology
2005
, vol. 
151
 (pg. 
373
-
383
)
167
Purevdorj
B
Costerton
JW
Stoodley
P
Influence of hydrodynamics and cell signaling on the structure and behavior of Pseudomonas aeruginosa biofilms
Appl. Environ. Microbiol.
2002
, vol. 
68
 (pg. 
4457
-
4464
)
168
Heydorn
A
Ersboll
B
Kato
J
Hentzer
M
Parsek
MR
Tolker-Nielsen
T
Givskov
M
Molin
S
Statistical analysis of Pseudomonas aeruginosa biofilm development: impact of mutations in genes involved in twitching motility, cell-to-cell signaling, and stationary-phase sigma factor expression
Appl. Environ. Microbiol.
2002
, vol. 
68
 (pg. 
2008
-
2017
)
169
Smith
EE
Buckley
DG
Wu
Z
Saenphimmachak
C
Hoffman
LR
D'Argenio
DA
Miller
SI
Ramsey
BW
Speert
DP
Moskowitz
SM
, et al. 
Genetic adaptation by Pseudomonas aeruginosa to the airways of cystic fibrosis patients
Proc. Natl Acad. Sci. USA
2006
, vol. 
103
 (pg. 
8487
-
8492
)
170
Van Delden
C
Pesci
EC
Pearson
JP
Iglewski
BH
Starvation selection restores elastase and rhamnolipid production in a Pseudomonas aeruginosa quorum-sensing mutant
Infect. Immun.
1998
, vol. 
66
 (pg. 
4499
-
4502
)
171
Kirisits
MJ
Parsek
MR
Does Pseudomonas aeruginosa use intercellular signalling to build biofilm communities?
Cell. Microbiol.
2006
, vol. 
8
 (pg. 
1841
-
1849
)
172
Lewis
K
Multidrug tolerance of biofilms and persister cells
Curr. Top. Microbiol. Immunol.
2008
, vol. 
322
 (pg. 
107
-
131
)
173
Keren
I
Kaldalu
N
Spoering
A
Wang
Y
Lewis
K
Persister cells and tolerance to antimicrobials
FEMS Microbiol. Lett.
2004
, vol. 
230
 (pg. 
13
-
18
)
174
Keren
I
Shah
D
Spoering
A
Kaldalu
N
Lewis
K
Specialized persister cells and the mechanism of multidrug tolerance in Escherichia coli
J. Bacteriol.
2004
, vol. 
186
 (pg. 
8172
-
8180
)
175
De Groote
VN
Verstraeten
N
Fauvart
M
Kint
CI
Verbeeck
AM
Beullens
S
Cornelis
P
Michiels
J
Novel persistence genes in Pseudomonas aeruginosa identified by high-throughput screening
FEMS Microbiol. Lett.
2009
, vol. 
297
 (pg. 
73
-
79
)
176
Fauvart
M
De Groote
VN
Michiels
J
Role of persister cells in chronic infections: clinical relevance and perspectives on anti-persister therapies
J. Med. Microbiol.
2011
, vol. 
60
 (pg. 
699
-
709
)
177
Lewis
K
Persister cells
Annu. Rev. Microbiol.
2010
, vol. 
64
 (pg. 
357
-
372
)
178
Pressler
T
Bohmova
C
Conway
S
Dumcius
S
Hjelte
L
Hoiby
N
Kollberg
H
Tummler
B
Vavrova
V
Chronic Pseudomonas aeruginosa infection definition: EuroCareCF Working Group report
J. Cyst. Fibros.
2011
, vol. 
10
 (pg. 
S75
-
S78
)
179
de Jong
PA
Nakano
Y
Lequin
MH
Mayo
JR
Woods
R
Pare
PD
Tiddens
HA
Progressive damage on high resolution computed tomography despite stable lung function in cystic fibrosis
Eur. Respir. J.
2004
, vol. 
23
 (pg. 
93
-
97
)
180
Meyer
KC
Sharma
A
Regional variability of lung inflammation in cystic fibrosis
Am. J. Respir. Crit. Care Med.
1997
, vol. 
156
 (pg. 
1536
-
1540
)
181
Tiddens
HA
Detecting early structural lung damage in cystic fibrosis
Pediatr. Pulmonol.
2002
, vol. 
34
 (pg. 
228
-
231
)
182
Worlitzsch
D
Tarran
R
Ulrich
M
Schwab
U
Cekici
A
Meyer
KC
Birrer
P
Bellon
G
Berger
J
Weiss
T
, et al. 
Effects of reduced mucus oxygen concentration in airway Pseudomonas infections of cystic fibrosis patients
J. Clin. Invest.
2002
, vol. 
109
 (pg. 
317
-
325
)
183
Doggett
RG
Harrison
GM
Carter
RE
Mucoid Pseudomonas aeruginosa in patients with chronic illnesses
Lancet
1971
, vol. 
1
 (pg. 
236
-
237
)
184
Hoiby
N
Pseudomonas aeruginosa infection in cystic fibrosis. Relationship between mucoid strains of Pseudomonas aeruginosa and the humoral immune response
Acta. Pathol. Microbiol. Scand. [B] Microbiol. Immunol.
1974
, vol. 
82
 (pg. 
551
-
558
)
185
Evans
LR
Linker
A
Production and characterization of the slime polysaccharide of Pseudomonas aeruginosa
J. Bacteriol.
1973
, vol. 
116
 (pg. 
915
-
924
)
186
Pedersen
SS
Espersen
F
Hoiby
N
Jensen
T
Immunoglobulin A and immunoglobulin G antibody responses to alginates from Pseudomonas aeruginosa in patients with cystic fibrosis
J. Clin. Microbiol.
1990
, vol. 
28
 (pg. 
747
-
755
)
187
Pier
GB
Coleman
F
Grout
M
Franklin
M
Ohman
DE
Role of alginate O acetylation in resistance of mucoid Pseudomonas aeruginosa to opsonic phagocytosis
Infect. Immun.
2001
, vol. 
69
 (pg. 
1895
-
1901
)
188
Simpson
JA
Smith
SE
Dean
RT
Alginate inhibition of the uptake of Pseudomonas aeruginosa by macrophages
J. Gen. Microbiol.
1988
, vol. 
134
 (pg. 
29
-
36
)
189
Mathee
K
Ciofu
O
Sternberg
C
Lindum
PW
Campbell
JI
Jensen
P
Johnsen
AH
Givskov
M
Ohman
DE
Molin
S
, et al. 
Mucoid conversion of Pseudomonas aeruginosa by hydrogen peroxide: a mechanism for virulence activation in the cystic fibrosis lung
Microbiology
1999
, vol. 
145
 (pg. 
1349
-
1357
)
190
Govan
JR
Fyfe
JA
Mucoid Pseudomonas aeruginosa and cystic fibrosis: resistance of the mucoid form to carbenicillin, flucloxacillin and tobramycin and the isolation of mucoid variants in vitro
J. Antimicrob. Chemother.
1978
, vol. 
4
 (pg. 
233
-
240
)
191
Speert
DP
Farmer
SW
Campbell
ME
Musser
JM
Selander
RK
Kuo
S
Conversion of Pseudomonas aeruginosa to the phenotype characteristic of strains from patients with cystic fibrosis
J. Clin. Microbiol.
1990
, vol. 
28
 (pg. 
188
-
194
)
192
Terry
JM
Pina
SE
Mattingly
SJ
Environmental conditions which influence mucoid conversion Pseudomonas aeruginosa PAO1
Infect. Immun.
1991
, vol. 
59
 (pg. 
471
-
477
)
193
DeVries
CA
Ohman
DE
Mucoid-to-nonmucoid conversion in alginate-producing Pseudomonas aeruginosa often results from spontaneous mutations in algT, encoding a putative alternate sigma factor, and shows evidence for autoregulation
J. Bacteriol.
1994
, vol. 
176
 (pg. 
6677
-
6687
)
194
Martin
DW
Holloway
BW
Deretic
V
Characterization of a locus determining the mucoid status of Pseudomonas aeruginosa: AlgU shows sequence similarities with a Bacillus sigma factor
J. Bacteriol.
1993
, vol. 
175
 (pg. 
1153
-
1164
)
195
Martin
DW
Schurr
MJ
Mudd
MH
Deretic
V
Differentiation of Pseudomonas aeruginosa into the alginate-producing form: inactivation of mucB causes conversion to mucoidy
Mol. Microbiol.
1993
, vol. 
9
 (pg. 
497
-
506
)
196
Mathee
K
McPherson
CJ
Ohman
DE
Posttranslational control of the algT (algU)-encoded sigma22 for expression of the alginate regulon in Pseudomonas aeruginosa and localization of its antagonist proteins MucA and MucB (AlgN)
J. Bacteriol.
1997
, vol. 
179
 (pg. 
3711
-
3720
)
197
Mathee
K
Kharazami
AA
Hoiby
N
McLean
RJC
Molecular Ecology of Biofilms
2002
Norfolk
Horizon
(pg. 
23
-
55
)
198
Goldberg
JB
Gorman
WL
Flynn
JL
Ohman
DE
A mutation in algN permits trans activation of alginate production by algT in Pseudomonas species
J. Bacteriol.
1993
, vol. 
175
 (pg. 
1303
-
1308
)
199
Martin
DW
Schurr
MJ
Mudd
MH
Govan
JR
Holloway
BW
Deretic
V
Mechanism of conversion to mucoidy in Pseudomonas aeruginosa infecting cystic fibrosis patients
Proc. Natl Acad. Sci. USA
1993
, vol. 
90
 (pg. 
8377
-
8381
)
200
Ohman
D
Rehm
BHA
Alginates: Biology and Applications
2009
, vol. 
Vol. 13
 
Berlin/Heidelberg
Springer
(pg. 
117
-
133
)
201
Damron
FH
Goldberg
JB
Proteolytic regulation of alginate overproduction in Pseudomonas aeruginosa
Mol. Microbiol.
2012
, vol. 
84
 (pg. 
595
-
607
)
202
Damron
FH
Yu
HD
Pseudomonas aeruginosa MucD regulates the alginate pathway through activation of MucA degradation via MucP proteolytic activity
J. Bacteriol.
2011
, vol. 
193
 (pg. 
286
-
291
)
203
Martin
DW
Schurr
MJ
Yu
H
Deretic
V
Analysis of promoters controlled by the putative sigma factor AlgU regulating conversion to mucoidy in Pseudomonas aeruginosa: relationship to sigma E and stress response
J. Bacteriol.
1994
, vol. 
176
 (pg. 
6688
-
6696
)
204
Wozniak
DJ
Ohman
DE
Transcriptional analysis of the Pseudomonas aeruginosa genes algR, algB, and algD reveals a hierarchy of alginate gene expression which is modulated by algT
J. Bacteriol.
1994
, vol. 
176
 (pg. 
6007
-
6014
)
205
Wozniak
DJ
Sprinkle
AB
Baynham
PJ
Control of Pseudomonas aeruginosa algZ expression by the alternative sigma factor AlgT
J. Bacteriol.
2003
, vol. 
185
 (pg. 
7297
-
7300
)
206
Chitnis
CE
Ohman
DE
Genetic analysis of the alginate biosynthetic gene cluster of Pseudomonas aeruginosa shows evidence of an operonic structure
Mol. Microbiol.
1993
, vol. 
8
 (pg. 
583
-
593
)
207
Deretic
V
Gill
JF
Chakrabarty
AM
Gene algD coding for GDPmannose dehydrogenase is transcriptionally activated in mucoid Pseudomonas aeruginosa
J. Bacteriol.
1987
, vol. 
169
 (pg. 
351
-
358
)
208
Leech
AJ
Sprinkle
A
Wood
L
Wozniak
DJ
Ohman
DE
The NtrC family regulator AlgB, which controls alginate biosynthesis in mucoid Pseudomonas aeruginosa, binds directly to the algD promoter
J. Bacteriol.
2008
, vol. 
190
 (pg. 
581
-
589
)
209
Kato
J
Chakrabarty
AM
Purification of the regulatory protein AlgR1 and its binding in the far upstream region of the algD promoter in Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
1991
, vol. 
88
 (pg. 
1760
-
1764
)
210
Mohr
CD
Martin
DW
Konyecsni
WM
Govan
JR
Lory
S
Deretic
V
Role of the far-upstream sites of the algD promoter and the algR and rpoN genes in environmental modulation of mucoidy in Pseudomonas aeruginosa
J. Bacteriol.
1990
, vol. 
172
 (pg. 
6576
-
6580
)
211
Baynham
PJ
Brown
AL
Hall
LL
Wozniak
DJ
Pseudomonas aeruginosa AlgZ, a ribbon-helix-helix DNA-binding protein, is essential for alginate synthesis and algD transcriptional activation
Mol. Microbiol.
1999
, vol. 
33
 (pg. 
1069
-
1080
)
212
Damron
FH
Qiu
D
Yu
HD
The Pseudomonas aeruginosa sensor kinase KinB negatively controls alginate production through AlgW-dependent MucA proteolysis
J. Bacteriol.
2009
, vol. 
191
 (pg. 
2285
-
2295
)
213
Wood
LF
Leech
AJ
Ohman
DE
Cell wall-inhibitory antibiotics activate the alginate biosynthesis operon in Pseudomonas aeruginosa: Roles of sigma (AlgT) and the AlgW and Prc proteases
Mol. Microbiol.
2006
, vol. 
62
 (pg. 
412
-
426
)
214
Ma
S
Selvaraj
U
Ohman
DE
Quarless
R
Hassett
DJ
Wozniak
DJ
Phosphorylation-independent activity of the response regulators AlgB and AlgR in promoting alginate biosynthesis in mucoid Pseudomonas aeruginosa
J. Bacteriol.
1998
, vol. 
180
 (pg. 
956
-
968
)
215
Zegans
ME
Wozniak
D
Griffin
E
Toutain-Kidd
CM
Hammond
JH
Garfoot
A
Lam
JS
Pseudomonas aeruginosa exopolysaccharide Psl promotes resistance to the biofilm inhibitor polysorbate 80
Antimicrob. Agents Chemother.
2012
, vol. 
56
 (pg. 
4112
-
4122
)
216
Fujiwara
S
Zielinski
NA
Chakrabarty
AM
Enhancer-like activity of A1gR1-binding site in alginate gene activation: positional, orientational, and sequence specificity
J. Bacteriol.
1993
, vol. 
175
 (pg. 
5452
-
5459
)
217
Ye
RW
Zielinski
NA
Chakrabarty
AM
Purification and characterization of phosphomannomutase/phosphoglucomutase from Pseudomonas aeruginosa involved in biosynthesis of both alginate and lipopolysaccharide
J. Bacteriol.
1994
, vol. 
176
 (pg. 
4851
-
4857
)
218
Zielinski
NA
Maharaj
R
Roychoudhury
S
Danganan
CE
Hendrickson
W
Chakrabarty
AM
Alginate synthesis in Pseudomonas aeruginosa: environmental regulation of the algC promoter
J. Bacteriol.
1992
, vol. 
174
 (pg. 
7680
-
7688
)
219
Morici
LA
Carterson
AJ
Wagner
VE
Frisk
A
Schurr
JR
Honer zu Bentrup
K
Hassett
DJ
Iglewski
BH
Sauer
K
Schurr
MJ
Pseudomonas aeruginosa AlgR represses the Rhl quorum-sensing system in a biofilm-specific manner
J. Bacteriol.
2007
, vol. 
189
 (pg. 
7752
-
7764
)
220
Belete
B
Lu
H
Wozniak
DJ
Pseudomonas aeruginosa AlgR regulates type IV pilus biosynthesis by activating transcription of the fimU-pilVWXY1Y2E operon
J. Bacteriol.
2008
, vol. 
190
 (pg. 
2023
-
2030
)
221
Lizewski
SE
Lundberg
DS
Schurr
MJ
The transcriptional regulator AlgR is essential for Pseudomonas aeruginosa pathogenesis
Infect. Immun.
2002
, vol. 
70
 (pg. 
6083
-
6093
)
222
Carterson
AJ
Morici
LA
Jackson
DW
Frisk
A
Lizewski
SE
Jupiter
R
Simpson
K
Kunz
DA
Davis
SH
Schurr
JR
, et al. 
The transcriptional regulator AlgR controls cyanide production in Pseudomonas aeruginosa
J. Bacteriol.
2004
, vol. 
186
 (pg. 
6837
-
6844
)
223
Cody
WL
Pritchett
CL
Jones
AK
Carterson
AJ
Jackson
D
Frisk
A
Wolfgang
MC
Schurr
MJ
Pseudomonas aeruginosa AlgR controls cyanide production in an AlgZ-dependent manner
J. Bacteriol.
2009
, vol. 
191
 (pg. 
2993
-
3002
)
224
Jones
AK
Fulcher
NB
Balzer
GJ
Urbanowski
ML
Pritchett
CL
Schurr
MJ
Yahr
TL
Wolfgang
MC
Activation of the Pseudomonas aeruginosa AlgU regulon through mucA mutation inhibits cyclic AMP/Vfr signaling
J. Bacteriol.
2010
, vol. 
192
 (pg. 
5709
-
5717
)
225
Lizewski
SE
Schurr
JR
Jackson
DW
Frisk
A
Carterson
AJ
Schurr
MJ
Identification of AlgR-regulated genes in Pseudomonas aeruginosa by use of microarray analysis
J. Bacteriol.
2004
, vol. 
186
 (pg. 
5672
-
5684
)
226
Remminghorst
U
Rehm
BH
Alg44, a unique protein required for alginate biosynthesis in Pseudomonas aeruginosa
FEBS Lett.
2006
, vol. 
580
 (pg. 
3883
-
3888
)
227
Hay
ID
Remminghorst
U
Rehm
BH
MucR, a novel membrane-associated regulator of alginate biosynthesis in Pseudomonas aeruginosa
Appl. Environ. Microbiol.
2009
, vol. 
75
 (pg. 
1110
-
1120
)
228
Merighi
M
Lee
VT
Hyodo
M
Hayakawa
Y
Lory
S
The second messenger bis-(3'-5')-cyclic-GMP and its PilZ domain-containing receptor Alg44 are required for alginate biosynthesis in Pseudomonas aeruginosa
Mol. Microbiol.
2007
, vol. 
65
 (pg. 
876
-
895
)
229
Ochsner
UA
Wilderman
PJ
Vasil
AI
Vasil
ML
GeneChip expression analysis of the iron starvation response in Pseudomonas aeruginosa: identification of novel pyoverdine biosynthesis genes
Mol. Microbiol.
2002
, vol. 
45
 (pg. 
1277
-
1287
)
230
Palma
M
Worgall
S
Quadri
LE
Transcriptome analysis of the Pseudomonas aeruginosa response to iron
Arch. Microbiol.
2003
, vol. 
180
 (pg. 
374
-
379
)
231
Visca
P
Imperi
F
Lamont
IL
Pyoverdine siderophores: from biogenesis to biosignificance
Trends Microbiol.
2007
, vol. 
15
 (pg. 
22
-
30
)
232
Poole
K
McKay
GA
Iron acquisition and its control in Pseudomonas aeruginosa: many roads lead to Rome
Front. Biosci.
2003
, vol. 
8
 (pg. 
d661
-
d686
)
233
Ochsner
UA
Johnson
Z
Vasil
ML
Genetics and regulation of two distinct haem-uptake systems, phu and has, in Pseudomonas aeruginosa
Microbiology
2000
, vol. 
146
 (pg. 
185
-
198
)
234
Touati
D
Iron and oxidative stress in bacteria
Arch. Biochem. Biophys.
2000
, vol. 
373
 (pg. 
1
-
6
)
235
Vasil
ML
Ochsner
UA
The response of Pseudomonas aeruginosa to iron: genetics, biochemistry and virulence
Mol. Microbiol.
1999
, vol. 
34
 (pg. 
399
-
413
)
236
Pohl
E
Haller
JC
Mijovilovich
A
Meyer-Klaucke
W
Garman
E
Vasil
ML
Architecture of a protein central to iron homeostasis: crystal structure and spectroscopic analysis of the ferric uptake regulator
Mol. Microbiol.
2003
, vol. 
47
 (pg. 
903
-
915
)
237
Wilderman
PJ
Sowa
NA
FitzGerald
DJ
FitzGerald
PC
Gottesman
S
Ochsner
UA
Vasil
ML
Identification of tandem duplicate regulatory small RNAs in Pseudomonas aeruginosa involved in iron homeostasis
Proc. Natl Acad. Sci. USA
2004
, vol. 
101
 (pg. 
9792
-
9797
)
238
Cornelis
P
Matthijs
S
Van Oeffelen
L
Iron uptake regulation in Pseudomonas aeruginosa
Biometals
2009
, vol. 
22
 (pg. 
15
-
22
)
239
Ochsner
UA
Vasil
ML
Gene repression by the ferric uptake regulator in Pseudomonas aeruginosa: cycle selection of iron-regulated genes
Proc. Natl Acad. Sci. USA
1996
, vol. 
93
 (pg. 
4409
-
4414
)
240
Wilderman
PJ
Vasil
AI
Johnson
Z
Wilson
MJ
Cunliffe
HE
Lamont
IL
Vasil
ML
Characterization of an endoprotease (PrpL) encoded by a PvdS-regulated gene in Pseudomonas aeruginosa
Infect. Immun.
2001
, vol. 
69
 (pg. 
5385
-
5394
)
241
Ochsner
UA
Johnson
Z
Lamont
IL
Cunliffe
HE
Vasil
ML
Exotoxin A production in Pseudomonas aeruginosa requires the iron-regulated pvdS gene encoding an alternative sigma factor
Mol. Microbiol.
1996
, vol. 
21
 (pg. 
1019
-
1028
)
242
Gaines
JM
Carty
NL
Tiburzi
F
Davinic
M
Visca
P
Colmer-Hamood
JA
Hamood
AN
Regulation of the Pseudomonas aeruginosa toxA, regA and ptxR genes by the iron-starvation sigma factor PvdS under reduced levels of oxygen
Microbiology
2007
, vol. 
153
 (pg. 
4219
-
4233
)
243
Xiong
YQ
Vasil
ML
Johnson
Z
Ochsner
UA
Bayer
AS
The oxygen- and iron-dependent sigma factor pvdS of Pseudomonas aeruginosa is an important virulence factor in experimental infective endocarditis
J. Infect. Dis.
2000
, vol. 
181
 (pg. 
1020
-
1026
)
244
Singh
PK
Parsek
MR
Greenberg
EP
Welsh
MJ
A component of innate immunity prevents bacterial biofilm development
Nature
2002
, vol. 
417
 (pg. 
552
-
555
)
245
Singh
PK
Iron sequestration by human lactoferrin stimulates P. aeruginosa surface motility and blocks biofilm formation
Biometals
2004
, vol. 
17
 (pg. 
267
-
270
)
246
Banin
E
Vasil
ML
Greenberg
EP
Iron and Pseudomonas aeruginosa biofilm formation
Proc. Natl Acad. Sci. USA
2005
, vol. 
102
 (pg. 
11076
-
11081
)
247
Yang
L
Barken
KB
Skindersoe
ME
Christensen
AB
Givskov
M
Tolker-Nielsen
T
Effects of iron on DNA release and biofilm development by Pseudomonas aeruginosa
Microbiology
2007
, vol. 
153
 (pg. 
1318
-
1328
)
248
Bollinger
N
Hassett
DJ
Iglewski
BH
Costerton
JW
McDermott
TR
Gene expression in Pseudomonas aeruginosa: evidence of iron override effects on quorum sensing and biofilm-specific gene regulation
J. Bacteriol.
2001
, vol. 
183
 (pg. 
1990
-
1996
)
249
Kim
EJ
Wang
W
Deckwer
WD
Zeng
AP
Expression of the quorum-sensing regulatory protein LasR is strongly affected by iron and oxygen concentrations in cultures of Pseudomonas aeruginosa irrespective of cell density
Microbiology
2005
, vol. 
151
 (pg. 
1127
-
1138
)
250
Schuster
M
Lostroh
CP
Ogi
T
Greenberg
EP
Identification, timing, and signal specificity of Pseudomonas aeruginosa quorum-controlled genes: a transcriptome analysis
J. Bacteriol.
2003
, vol. 
185
 (pg. 
2066
-
2079
)
251
Juhas
M
Wiehlmann
L
Salunkhe
P
Lauber
J
Buer
J
Tummler
B
GeneChip expression analysis of the VqsR regulon of Pseudomonas aeruginosa TB
FEMS Microbiol. Lett.
2005
, vol. 
242
 (pg. 
287
-
295
)
252
Visca
P
Leoni
L
Wilson
MJ
Lamont
IL
Iron transport and regulation, cell signalling and genomics: lessons from Escherichia coli and Pseudomonas
Mol. Microbiol.
2002
, vol. 
45
 (pg. 
1177
-
1190
)
253
Hazan
R
He
J
Xiao
G
Dekimpe
V
Apidianakis
Y
Lesic
B
Astrakas
C
Deziel
E
Lepine
F
Rahme
LG
Homeostatic interplay between bacterial cell-cell signaling and iron in virulence
PLoS Pathog.
2010
, vol. 
6
 pg. 
e1000810
 
254
Oglesby
AG
Farrow
JM
3rd
Lee
JH
Tomaras
AP
Greenberg
EP
Pesci
EC
Vasil
ML
The influence of iron on Pseudomonas aeruginosa physiology: a regulatory link between iron and quorum sensing
J. Biol. Chem.
2008
, vol. 
283
 (pg. 
15558
-
15567
)
255
Bleves
S
Viarre
V
Salacha
R
Michel
GP
Filloux
A
Voulhoux
R
Protein secretion systems in Pseudomonas aeruginosa: a wealth of pathogenic weapons
Int. J. Med. Microbiol.
2010
, vol. 
300
 (pg. 
534
-
543
)
256
Hauser
AR
The type III secretion system of Pseudomonas aeruginosa: infection by injection
Nat. Rev. Microbiol.
2009
, vol. 
7
 (pg. 
654
-
665
)
257
Guzzo
J
Pages
JM
Duong
F
Lazdunski
A
Murgier
M
Pseudomonas aeruginosa alkaline protease: evidence for secretion genes and study of secretion mechanism
J. Bacteriol.
1991
, vol. 
173
 (pg. 
5290
-
5297
)
258
Matsumoto
K
Role of bacterial proteases in pseudomonal and serratial keratitis
Biol. Chem.
2004
, vol. 
385
 (pg. 
1007
-
1016
)
259
Wandersman
C
Delepelaire
P
Bacterial iron sources: from siderophores to hemophores
Annu. Rev. Microbiol.
2004
, vol. 
58
 (pg. 
611
-
647
)
260
Hood
RD
Singh
P
Hsu
F
Guvener
T
Carl
MA
Trinidad
RR
Silverman
JM
Ohlson
BB
Hicks
KG
Plemel
RL
, et al. 
A type VI secretion system of Pseudomonas aeruginosa targets a toxin to bacteria
Cell Host Microbe
2010
, vol. 
7
 (pg. 
25
-
37
)
261
Moscoso
JA
Mikkelsen
H
Heeb
S
Williams
P
Filloux
A
The Pseudomonas aeruginosa sensor RetS switches type III and type VI secretion via c-di-GMP signalling
Environ. Microbiol.
2011
, vol. 
13
 (pg. 
3128
-
3138
)
262
Turner
KH
Vallet-Gely
I
Dove
SL
Epigenetic control of virulence gene expression in Pseudomonas aeruginosa by a LysR-type transcription regulator
PLoS Genet.
2009
, vol. 
5
 pg. 
e1000779
 
263
Pearson
JP
Pesci
EC
Iglewski
BH
Roles of Pseudomonas aeruginosa las and rhl quorum-sensing systems in control of elastase and rhamnolipid biosynthesis genes
J. Bacteriol.
1997
, vol. 
179
 (pg. 
5756
-
5767
)
264
Chapon-Herve
V
Akrim
M
Latifi
A
Williams
P
Lazdunski
A
Bally
M
Regulation of the xcp secretion pathway by multiple quorum-sensing modulons in Pseudomonas aeruginosa
Mol. Microbiol.
1997
, vol. 
24
 (pg. 
1169
-
1178
)
265
Lazdunski
AM
Ventre
I
Sturgis
JN
Regulatory circuits and communication in Gram-negative bacteria
Nat. Rev. Microbiol.
2004
, vol. 
2
 (pg. 
581
-
592
)
266
Filloux
A
Bally
M
Soscia
C
Murgier
M
Lazdunski
A
Phosphate regulation in Pseudomonas aeruginosa: cloning of the alkaline phosphatase gene and identification of phoB- and phoR-like genes
Mol. Gen. Genet.
1988
, vol. 
212
 (pg. 
510
-
513
)
267
Ball
G
Durand
E
Lazdunski
A
Filloux
A
A novel type II secretion system in Pseudomonas aeruginosa
Mol. Microbiol.
2002
, vol. 
43
 (pg. 
475
-
485
)
268
Llamas
MA
van der Sar
A
Chu
BC
Sparrius
M
Vogel
HJ
Bitter
W
A novel extracytoplasmic function (ECF) sigma factor regulates virulence in Pseudomonas aeruginosa
PLoS Pathog.
2009
, vol. 
5
 pg. 
e1000572
 
269
Lesic
B
Starkey
M
He
J
Hazan
R
Rahme
LG
Quorum sensing differentially regulates Pseudomonas aeruginosa type VI secretion locus I and homologous loci II and III, which are required for pathogenesis
Microbiology
2009
, vol. 
155
 (pg. 
2845
-
2855
)
270
Mougous
JD
Cuff
ME
Raunser
S
Shen
A
Zhou
M
Gifford
CA
Goodman
AL
Joachimiak
G
Ordonez
CL
Lory
S
, et al. 
A virulence locus of Pseudomonas aeruginosa encodes a protein secretion apparatus
Science
2006
, vol. 
312
 (pg. 
1526
-
1530
)
271
Yahr
TL
Wolfgang
MC
Transcriptional regulation of the Pseudomonas aeruginosa type III secretion system
Mol. Microbiol.
2006
, vol. 
62
 (pg. 
631
-
640
)
272
Frank
DW
The exoenzyme S regulon of Pseudomonas aeruginosa
Mol. Microbiol.
1997
, vol. 
26
 (pg. 
621
-
629
)
273
Vallis
AJ
Yahr
TL
Barbieri
JT
Frank
DW
Regulation of ExoS production and secretion by Pseudomonas aeruginosa in response to tissue culture conditions
Infect. Immun.
1999
, vol. 
67
 (pg. 
914
-
920
)
274
Shen
DK
Filopon
D
Kuhn
L
Polack
B
Toussaint
B
PsrA is a positive transcriptional regulator of the type III secretion system in Pseudomonas aeruginosa
Infect. Immun.
2006
, vol. 
74
 (pg. 
1121
-
1129
)
275
Hogardt
M
Roeder
M
Schreff
AM
Eberl
L
Heesemann
J
Expression of Pseudomonas aeruginosa exoS is controlled by quorum sensing and RpoS
Microbiology
2004
, vol. 
150
 (pg. 
843
-
851
)
276
Dasgupta
N
Lykken
GL
Wolfgang
MC
Yahr
TL
A novel anti-anti-activator mechanism regulates expression of the Pseudomonas aeruginosa type III secretion system
Mol. Microbiol.
2004
, vol. 
53
 (pg. 
297
-
308
)
277
Wu
W
Jin
S
PtrB of Pseudomonas aeruginosa suppresses the type III secretion system under the stress of DNA damage
J. Bacteriol.
2005
, vol. 
187
 (pg. 
6058
-
6068
)
278
Hornef
MW
Roggenkamp
A
Geiger
AM
Hogardt
M
Jacobi
CA
Heesemann
J
Triggering the ExoS regulon of Pseudomonas aeruginosa: a GFP-reporter analysis of exoenzyme (Exo) S, ExoT and ExoU synthesis
Microb. Pathog.
2000
, vol. 
29
 (pg. 
329
-
343
)
279
Rietsch
A
Mekalanos
JJ
Metabolic regulation of type III secretion gene expression in Pseudomonas aeruginosa
Mol. Microbiol.
2006
, vol. 
59
 (pg. 
807
-
820
)
280
Dacheux
D
Epaulard
O
de Groot
A
Guery
B
Leberre
R
Attree
I
Polack
B
Toussaint
B
Activation of the Pseudomonas aeruginosa type III secretion system requires an intact pyruvate dehydrogenase aceAB operon
Infect. Immun.
2002
, vol. 
70
 (pg. 
3973
-
3977
)
281
Wu
W
Badrane
H
Arora
S
Baker
HV
Jin
S
MucA-mediated coordination of type III secretion and alginate synthesis in Pseudomonas aeruginosa
J. Bacteriol.
2004
, vol. 
186
 (pg. 
7575
-
7585
)
282
Jin
Y
Yang
H
Qiao
M
Jun
S
MexT regulates the type III secretion system through MexS and PtrC in Pseudomonas aeruginosa
J. Bacteriol.
2011
, vol. 
193
 (pg. 
399
-
410
)
283
Mulcahy
H
O'Callaghan
J
O'Grady
EP
Adams
C
O'Gara
F
The posttranscriptional regulator RsmA plays a role in the interaction between Pseudomonas aeruginosa and human airway epithelial cells by positively regulating the type III secretion system
Infect. Immun.
2006
, vol. 
74
 (pg. 
3012
-
3015
)
284
O'Callaghan
J
Reen
FJ
Adams
C
O'Gara
F
Low oxygen induces the type III secretion system in Pseudomonas aeruginosa via modulation of the small RNAs rsmZ and rsmY
Microbiology
2011
, vol. 
157
 (pg. 
3417
-
3428
)
285
Dubnau
D
Losick
R
Bistability in bacteria
Mol. Microbiol.
2006
, vol. 
61
 (pg. 
564
-
572
)
286
Storz
G
Altuvia
S
Wassarman
KM
An abundance of RNA regulators
Annu. Rev. Biochem.
2005
, vol. 
74
 (pg. 
199
-
217
)
287
Papenfort
K
Vogel
J
Multiple target regulation by small noncoding RNAs rewires gene expression at the post-transcriptional level
Res. Microbiol.
2009
, vol. 
160
 (pg. 
278
-
287
)
288
Gottesman
S
The small RNA regulators of Escherichia coli: roles and mechanisms
Annu. Rev. Microbiol.
2004
, vol. 
58
 (pg. 
303
-
328
)
289
Ferrara
S
Brugnoli
M
De Bonis
A
Righetti
F
Delvillani
F
Deho
G
Horner
D
Briani
F
Bertoni
G
Comparative profiling of Pseudomonas aeruginosa strains reveals differential expression of novel unique and conserved small RNAs
PLoS One
2012
, vol. 
7
 pg. 
e36553
 
290
Heurlier
K
Williams
F
Heeb
S
Dormond
C
Pessi
G
Singer
D
Camara
M
Williams
P
Haas
D
Positive control of swarming, rhamnolipid synthesis, and lipase production by the posttranscriptional RsmA/RsmZ system in Pseudomonas aeruginosa PAO1
J. Bacteriol.
2004
, vol. 
186
 (pg. 
2936
-
2945
)
291
Heeb
S
Blumer
C
Haas
D
Regulatory RNA as mediator in GacA/RsmA-dependent global control of exoproduct formation in Pseudomonas fluorescens CHA0
J. Bacteriol.
2002
, vol. 
184
 (pg. 
1046
-
1056
)
292
Valverde
C
Heeb
S
Keel
C
Haas
D
RsmY, a small regulatory RNA, is required in concert with RsmZ for GacA-dependent expression of biocontrol traits in Pseudomonas fluorescens CHA0
Mol. Microbiol.
2003
, vol. 
50
 (pg. 
1361
-
1379
)
293
Kay
E
Dubuis
C
Haas
D
Three small RNAs jointly ensure secondary metabolism and biocontrol in Pseudomonas fluorescens CHA0
Proc. Natl Acad. Sci. USA
2005
, vol. 
102
 (pg. 
17136
-
17141
)
294
Heeb
S
Kuehne
SA
Bycroft
M
Crivii
S
Allen
MD
Haas
D
Camara
M
Williams
P
Functional analysis of the post-transcriptional regulator RsmA reveals a novel RNA-binding site
J. Mol. Biol.
2006
, vol. 
355
 (pg. 
1026
-
1036
)
295
Kay
E
Humair
B
Denervaud
V
Riedel
K
Spahr
S
Eberl
L
Valverde
C
Haas
D
Two GacA-dependent small RNAs modulate the quorum-sensing response in Pseudomonas aeruginosa
J. Bacteriol.
2006
, vol. 
188
 (pg. 
6026
-
6033
)
296
Bordi
C
Lamy
MC
Ventre
I
Termine
E
Hachani
A
Fillet
S
Roche
B
Bleves
S
Mejean
V
Lazdunski
A
, et al. 
Regulatory RNAs and the HptB/RetS signalling pathways fine-tune Pseudomonas aeruginosa pathogenesis
Mol. Microbiol.
2010
, vol. 
76
 (pg. 
1427
-
1443
)
297
Hsu
JL
Chen
HC
Peng
HL
Chang
HY
Characterization of the histidine-containing phosphotransfer protein B-mediated multistep phosphorelay system in Pseudomonas aeruginosa PAO1
J. Biol. Chem.
2008
, vol. 
283
 (pg. 
9933
-
9944
)
298
Petrova
OE
Sauer
K
The novel two-component regulatory system BfiSR regulates biofilm development by controlling the small RNA rsmZ through CafA
J. Bacteriol.
2010
, vol. 
192
 (pg. 
5275
-
5288
)
299
Brencic
A
McFarland
KA
McManus
HR
Castang
S
Mogno
I
Dove
SL
Lory
S
The GacS/GacA signal transduction system of Pseudomonas aeruginosa acts exclusively through its control over the transcription of the RsmY and RsmZ regulatory small RNAs
Mol. Microbiol.
2009
, vol. 
73
 (pg. 
434
-
445
)
300
Lapouge
K
Schubert
M
Allain
FH
Haas
D
Gac/Rsm signal transduction pathway of gamma-proteobacteria: from RNA recognition to regulation of social behaviour
Mol. Microbiol.
2008
, vol. 
67
 (pg. 
241
-
253
)
301
Sorger-Domenigg
T
Sonnleitner
E
Kaberdin
VR
Blasi
U
Distinct and overlapping binding sites of Pseudomonas aeruginosa Hfq and RsmA proteins on the non-coding RNA RsmY
Biochem. Biophys. Res. Commun.
2007
, vol. 
352
 (pg. 
769
-
773
)
302
Moreno
R
Marzi
S
Romby
P
Rojo
F
The Crc global regulator binds to an unpaired A-rich motif at the Pseudomonas putida alkS mRNA coding sequence and inhibits translation initiation
Nucleic Acids Res.
2009
, vol. 
37
 (pg. 
7678
-
7690
)
303
Moreno
R
Rojo
F
The target for the Pseudomonas putida Crc global regulator in the benzoate degradation pathway is the BenR transcriptional regulator
J. Bacteriol.
2008
, vol. 
190
 (pg. 
1539
-
1545
)
304
Moreno
R
Ruiz-Manzano
A
Yuste
L
Rojo
F
The Pseudomonas putida Crc global regulator is an RNA binding protein that inhibits translation of the AlkS transcriptional regulator
Mol. Microbiol.
2007
, vol. 
64
 (pg. 
665
-
675
)
305
Sonnleitner
E
Abdou
L
Haas
D
Small RNA as global regulator of carbon catabolite repression in Pseudomonas aeruginosa
Proc. Natl Acad. Sci. USA
2009
, vol. 
106
 (pg. 
21866
-
21871
)
306
Yeung
AT
Bains
M
Hancock
RE
The sensor kinase CbrA is a global regulator that modulates metabolism, virulence, and antibiotic resistance in Pseudomonas aeruginosa
J. Bacteriol.
2011
, vol. 
193
 (pg. 
918
-
931
)
307
Abdou
L
Chou
HT
Haas
D
Lu
CD
Promoter recognition and activation by the global response regulator CbrB in Pseudomonas aeruginosa
J. Bacteriol.
2011
, vol. 
193
 (pg. 
2784
-
2792
)
308
Li
W
Lu
CD
Regulation of carbon and nitrogen utilization by CbrAB and NtrBC two-component systems in Pseudomonas aeruginosa
J. Bacteriol.
2007
, vol. 
189
 (pg. 
5413
-
5420
)
309
Nishijyo
T
Haas
D
Itoh
Y
The CbrA-CbrB two-component regulatory system controls the utilization of multiple carbon and nitrogen sources in Pseudomonas aeruginosa
Mol. Microbiol.
2001
, vol. 
40
 (pg. 
917
-
931
)
310
Rietsch
A
Wolfgang
MC
Mekalanos
JJ
Effect of metabolic imbalance on expression of type III secretion genes in Pseudomonas aeruginosa
Infect. Immun.
2004
, vol. 
72
 (pg. 
1383
-
1390
)
311
Linares
JF
Moreno
R
Fajardo
A
Martinez-Solano
L
Escalante
R
Rojo
F
Martinez
JL
The global regulator Crc modulates metabolism, susceptibility to antibiotics and virulence in Pseudomonas aeruginosa
Environ. Microbiol.
2010
, vol. 
12
 (pg. 
3196
-
3212
)
312
O'Toole
GA
Gibbs
KA
Hager
PW
Phibbs
PV
Jr
Kolter
R
The global carbon metabolism regulator Crc is a component of a signal transduction pathway required for biofilm development by Pseudomonas aeruginosa
J. Bacteriol.
2000
, vol. 
182
 (pg. 
425
-
431
)
313
Sonnleitner
E
Romeo
A
Blasi
U
Small regulatory RNAs in Pseudomonas aeruginosa
RNA Biol.
2012
, vol. 
9
 (pg. 
364
-
371
)
314
Sonnleitner
E
Gonzalez
N
Sorger-Domenigg
T
Heeb
S
Richter
AS
Backofen
R
Williams
P
Huttenhofer
A
Haas
D
Blasi
U
The small RNA PhrS stimulates synthesis of the Pseudomonas aeruginosa quinolone signal
Mol. Microbiol.
2011
, vol. 
80
 (pg. 
868
-
885
)
315
Moll
I
Afonyushkin
T
Vytvytska
O
Kaberdin
VR
Blasi
U
Coincident Hfq binding and RNase E cleavage sites on mRNA and small regulatory RNAs
RNA
2003
, vol. 
9
 (pg. 
1308
-
1314
)
316
Masse
E
Gottesman
S
A small RNA regulates the expression of genes involved in iron metabolism in Escherichia coli
Proc. Natl Acad. Sci. USA
2002
, vol. 
99
 (pg. 
4620
-
4625
)
317
Vasil
ML
How we learnt about iron acquisition in Pseudomonas aeruginosa: a series of very fortunate events
Biometals
2007
, vol. 
20
 (pg. 
587
-
601
)
318
Oglesby-Sherrouse
AG
Vasil
ML
Characterization of a heme-regulated non-coding RNA encoded by the prrF locus of Pseudomonas aeruginosa
PLoS One
2010
, vol. 
5
 pg. 
e9930
 
319
Bornholdt
S
Boolean network models of cellular regulation: prospects and limitations
J. R. Soc. Interface
2008
, vol. 
5
 (pg. 
S85
-
S94
)
320
Kauffman
SA
Metabolic stability and epigenesis in randomly constructed genetic nets
J. Theor. Biol.
1969
, vol. 
22
 (pg. 
437
-
467
)
321
Thomas
R
Boolean formalization of genetic control circuits
J. Theor. Biol.
1973
, vol. 
42
 (pg. 
563
-
585
)
322
Shmulevich
I
Dougherty
ER
Zhang
W
Gene perturbation and intervention in probabilistic Boolean networks
Bioinformatics
2002
, vol. 
18
 (pg. 
1319
-
1331
)
323
Shmulevich
I
Dougherty
ER
Kim
S
Zhang
W
Probabilistic Boolean Networks: a rule-based uncertainty model for gene regulatory networks
Bioinformatics
2002
, vol. 
18
 (pg. 
261
-
274
)
324
Liang
J
Han
J
Stochastic Boolean networks: an efficient approach to modeling gene regulatory networks
BMC Syst. Biol.
2012
, vol. 
6
 pg. 
113
 
325
Babu
MM
Early Career Research Award Lecture. Structure, evolution and dynamics of transcriptional regulatory networks
Biochem. Soc. Trans.
2010
, vol. 
38
 (pg. 
1155
-
1178
)
326
Babu
MM
Lang
B
Aravind
L
Methods to reconstruct and compare transcriptional regulatory networks
Methods Mol. Biol.
2009
, vol. 
541
 (pg. 
163
-
180
)
327
Oestreicher
C
A history of chaos theory
Dialogues Clin. Neurosci.
2007
, vol. 
9
 (pg. 
279
-
289
)
328
Goldbeter
A
Biochemical Oscillations and Cellular Rythms
1996
Cambridge, UK
Cambridge University Press
329
Novak
B
Tyson
JJ
Design principles of biochemical oscillators
Nat. Rev. Mol. Cell. Biol.
2008
, vol. 
9
 (pg. 
981
-
991
)
330
Leloup
JC
Goldbeter
A
Chaos and birhythmicity in a model for circadian oscillations of the PER and TIM proteins in drosophila
J. Theor. Biol.
1999
, vol. 
198
 (pg. 
445
-
459
)
331
Suguna
C
Chowdhury
KK
Sinha
S
Minimal model for complex dynamics in cellular processes
Phys. Rev. E Stat. Phys. Plasmas Fluids Relat. Interdiscip. Topics
1999
, vol. 
60
 (pg. 
5943
-
5949
)
332
Zhang
Z
Ye
W
Qian
Y
Zheng
Z
Huang
X
Hu
G
Chaotic motifs in gene regulatory networks
PLoS One
2012
, vol. 
7
 pg. 
e39355
 
333
Trosko
JE
Ruch
RJ
Cell-cell communication in carcinogenesis
Front Biosci.
1998
, vol. 
3
 (pg. 
d208
-
d236
)
334
Thomas
R
D'Ari
R
Biological Feedback
1990
Boca Raton, FL
CRC Press
335
Ng
WL
Perez
L
Cong
J
Semmelhack
MF
Bassler
BL
Broad spectrum pro-quorum-sensing molecules as inhibitors of virulence in vibrios
PLoS Pathog.
2012
, vol. 
8
 pg. 
e1002767
 
336
Wiedenheft
B
Sternberg
SH
Doudna
JA
RNA-guided genetic silencing systems in bacteria and archaea
Nature
2012
, vol. 
482
 (pg. 
331
-
338
)
337
Sonnleitner
E
Sorger-Domenigg
T
Madej
MJ
Findeiss
S
Hackermuller
J
Huttenhofer
A
Stadler
PF
Blasi
U
Moll
I
Detection of small RNAs in Pseudomonas aeruginosa by RNomics and structure-based bioinformatic tools
Microbiology
2008
, vol. 
154
 (pg. 
3175
-
3187
)
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by-nc/3.0/), which permits non-commercial reuse, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com.

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.