Skip to main content
Advertisement
  • Loading metrics

Crosstalk between the serine/threonine kinase StkP and the response regulator ComE controls the stress response and intracellular survival of Streptococcus pneumoniae

  • Germán E. Piñas,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Departamento de Bioquímica Clínica—CIBICI (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina, Department of Biology, University of Utah, Salt Lake City, Utah, United States of America

  • Nicolás M. Reinoso-Vizcaino,

    Roles Conceptualization, Formal analysis, Investigation, Methodology, Validation, Visualization

    Affiliation Departamento de Bioquímica Clínica—CIBICI (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina

  • Nubia Y. Yandar Barahona,

    Roles Formal analysis, Investigation, Methodology, Visualization

    Affiliation Departamento de Bioquímica Clínica—CIBICI (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina

  • Paulo R. Cortes,

    Roles Formal analysis, Investigation, Methodology, Visualization

    Affiliation Departamento de Bioquímica Clínica—CIBICI (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina

  • Rosario Duran,

    Roles Formal analysis, Investigation, Methodology, Visualization

    Affiliation Instituto Pasteur de Montevideo and Instituto de Investigaciones Biológicas Clemente Estable, Unidad de Bioquímica y Proteómica Analíticas, Montevideo, Uruguay

  • Chandan Badapanda,

    Roles Data curation, Formal analysis, Investigation, Methodology, Software, Visualization

    Affiliation Bioinformatics Division, Xcelris Lab Limited, Ahmedabad, India

  • Ankita Rathore,

    Roles Formal analysis, Investigation, Methodology, Software, Validation, Visualization

    Affiliation Bioinformatics Division, Xcelris Lab Limited, Ahmedabad, India

  • Dario R. Bichara,

    Roles Conceptualization, Formal analysis, Investigation, Software, Visualization, Writing – original draft

    Affiliation Fundacion Instituto Leloir, CONICET, Buenos Aires, Argentina

  • Melina B. Cian,

    Roles Formal analysis

    Affiliation Departamento de Bioquímica Clínica—CIBICI (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina

  • Nadia B. Olivero,

    Roles Formal analysis, Investigation, Methodology

    Affiliation Departamento de Bioquímica Clínica—CIBICI (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina

  • Daniel R. Perez,

    Roles Conceptualization, Funding acquisition, Resources, Writing – original draft, Writing – review & editing

    Affiliation Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America

  • José Echenique

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Visualization, Writing – original draft, Writing – review & editing

    jeche@fcq.unc.edu.ar

    Affiliation Departamento de Bioquímica Clínica—CIBICI (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina

Abstract

Streptococcus pneumoniae is an opportunistic human bacterial pathogen that usually colonizes the upper respiratory tract, but the invasion and survival mechanism in respiratory epithelial cells remains elusive. Previously, we described that acidic stress-induced lysis (ASIL) and intracellular survival are controlled by ComE through a yet unknown activation mechanism under acidic conditions, which is independent of the ComD histidine kinase that activates this response regulator for competence development at pH 7.8. Here, we demonstrate that the serine/threonine kinase StkP is essential for ASIL, and show that StkP phosphorylates ComE at Thr128. Molecular dynamic simulations predicted that Thr128-phosphorylation induces conformational changes on ComE’s DNA-binding domain. Using nonphosphorylatable (ComET128A) and phosphomimetic (ComET128E) proteins, we confirmed that Thr128-phosphorylation increased the DNA-binding affinity of ComE. The non-phosphorylated form of ComE interacted more strongly with StkP than the phosphomimetic form at acidic pH, suggesting that pH facilitated crosstalk. To identify the ComE-regulated genes under acidic conditions, a comparative transcriptomic analysis was performed between the comET128A and wt strains, and differential expression of 104 genes involved in different cellular processes was detected, suggesting that the StkP/ComE pathway induced global changes in response to acidic stress. In the comET128A mutant, the repression of spxB and sodA correlated with decreased H2O2 production, whereas the reduced expression of murN correlated with an increased resistance to cell wall antibiotic-induced lysis, compatible with cell wall alterations. In the comET128A mutant, ASIL was blocked and acid tolerance response was higher compared to the wt strain. These phenotypes, accompanied with low H2O2 production, are likely responsible for the increased survival in pneumocytes of the comET128A mutant. We propose that the StkP/ComE pathway controls the stress response, thus affecting the intracellular survival of S. pneumoniae in pneumocytes, one of the first barriers that this pathogen must cross to establish an infection.

Author summary

Streptococcus pneumoniae is a major human pathogen and is the causal agent of otitis (media) and sinusitis. It is also responsible for severe infections such as bacteremia, pneumonia, and meningitis, associated with 2 million annual deaths. Although this bacterium is part of the human nasopharynx commensal microbiota, it can become a pathogen and cross the epithelial cell barrier to establishing infections of varying intensity. Although S. pneumoniae is considered to be a typical extracellular pathogen, transient intracellular life forms have been found in eukaryotic cells, suggesting a putative survival mechanism. Here, we report that the serine-threonine kinase StkP was able to phosphorylate the response regulator ComE to control different cellular processes in response to environmental stress. Moreover, the phosphorylation of ComE on Thr128, and the consequent conformational and functional changes resulting from this event, extended the current knowledge of molecular activation mechanisms of response regulators. In this report, we provide evidence for the regulatory control exerted by the StkP/ComE pathway on acid-induced autolysis (associated with pneumolysin release), the acid tolerance response, and H2O2 production to modulate tissue damage and intracellular survival, which are ultimately linked to pneumococcal pathogenesis.

Introduction

Sensing and transducing external (or internal) signals into an appropriate physiological response is part of a microorganism strategy to survive in a constantly changing environment. Signal transduction is mainly carried out by protein kinases, which autophosphorylate upon sensing stimuli and then catalyze the phosphorylation of a specific substrate that initiates an adaptive cellular response. In prokaryotes, signaling pathways are mainly mediated by two-component systems (TCS) consisting of sensor histidine kinases (HK) that phosphorylate response regulators (RR) on a receiver domain, thereby activating the effector domains of these regulators to induce a physiological event in bacterial cells. Generally, the RR effector domains bind regions of DNA that control gene expression in response to environmental changes [1,2]. Each particular HK presents a remarkable specificity for its cognate RR and is capable of identifying particular RRs.

Eukaryotic-like Ser/Thr protein kinases (STKs) are also present in prokaryotes, where they play key roles in several cellular processes, including the central or secondary metabolism, developmental processes, cell division and virulence [3]. The major human bacterial pathogen Streptococcus pneumoniae (S. pneumoniae or the pneumococcus) encodes a single copy of StkP, a eukaryotic-like serine/threonine protein kinase gene [4]. StkP is a membrane protein composed of an N-terminal kinase domain facing the cytoplasm, a short transmembrane region, and an extracellular C-terminal region containing four PASTA (Penicillin-binding protein and Ser/Thr protein kinase Associated) domains [46]. Comparison of the global expression profile of the wild-type and ΔstkP strains has revealed that the transcription of genes involved in the cell wall metabolism, pyrimidine biosynthesis, DNA repair, iron uptake, and oxidative stress response are controlled by StkP, which explain why stkP mutations have pleiotropic effects [7]. It has also been described that StkP phosphorylates several target proteins, mainly on threonine residues, with PASTA domains being essential for kinase activity [810]. However, phosphorylation on serine residues seems to be independent of StkP [11]. Immunofluorescence microscopy of pneumococcal cells localized StkP to the cell division apparatus [12], with phenotypic studies having demonstrated its impact on several cellular functions [13,14]. In fact, the stkP mutant displayed morphological and growth defects, cell division alterations, increased LytA-dependent autolysis (induced by either antibiotics or growth at an alkaline pH of 7.8), reduced tolerance to stress conditions (including acidic stress), and pilus-mediated adherence in endothelial cells [4,7,11,1517]. StkP is also essential for virulence, being necessary for lung infection and for invading and growing in the bloodstream of intranasally infected mice [4].

In S. pneumoniae, transient competence development (ability to take up exogenous DNA) in exponentially growing cells is considered a stress response to alkaline pH [2], with its core regulatory circuit being controlled by the TCS ComDE. In this quorum sensing system, the membrane-integrated HK ComD senses the extracellular accumulation of a 17 amino acid competence stimulating peptide (CSP)[2]. Upon activation by a critical concentration of CSP, ComD phosphorylates the response regulator ComE at Asp58 [18], which consequently initiates the transcription of comCDE, comAB, and comX (a gene encoding an alternative sigma factor) [19,20]. ComX turns on the transcription of genes whose products are involved in DNA binding, uptake, and recombination [21]. In this sense, the competence development is considered to be a type of stress response to alkaline pH [2]. It has been reported that StkP can also regulate competence at pH 7.8. Cells lacking StkP do not develop natural competence [4] and show severely reduced CSP-induced competence [7,22], despite having increased expression of many genes of the CSP-regulated competence regulon [7].

To invade tissues, S. pneumoniae must overcome a variety of stress situations, such as acidic pH, as a consequence of host inflammatory responses against the invading pathogen [23]. This characteristic local acidosis is caused by infiltration of neutrophils and activation of inflammatory cells, which leads to increased energy and oxygen demand, accelerated glucose consumption via glycolysis and thus increased lactic acid secretion [2426]. For instance, pH values obtained from pleural fluids from patients with acute bacterial pneumonia caused by S. pneumoniae showed an acidic pH close to 6.80 [27]. Interestingly, the lowest pH value that S. pneumoniae has been shown to be tolerant to is around 4.4 in phagosomal vesicles during the first few minutes after phagocytosis [28]. Although S. pneumoniae is considered a typical extracellular pathogen, a transient intracellular life was described, suggesting that it can survive inside eukaryotic cells. S. pneumoniae can cross brain microvascular endothelial cells inside vesicles derived from early and/or late endosomes [29] [30]. It is well accepted that acidification is essential to endosome/lysosome maturation, with early endosomes having a pH in the 6.8–6.1 range, late endosomes in the 6.0–4.8 range, whereas lysosomal pH values can drop to 4.5 [31]. In the case of a putative endosomal survival, S. pneumoniae must survive acidic conditions. Martin-Galiano et al [32] described that S. pneumoniae is able to induce an acid tolerance response (ATR) mechanism. Previously, we also showed that F0.F1-ATPase, a proton pump that controls intracellular pH, is relevant for ATR induction in S. pneumoniae. In addition, we demonstrated that the F0.F1-ATPase and ATR are necessary for the intracellular survival of the pneumococcus in macrophages [33].

As part of the acidic stress response, we have reported that exposure of S. pneumoniae to acidic culture conditions triggers a lytic response by the major autolysin LytA. The acidic-stress induced lysis (ASIL) response is promoted by ComE and repressed by the CiaRH TCS. Despite requiring ComE, ASIL does not depend on CSP or ComD. Curiously, the comE gene is induced by acidic stress, but the competence-related ComX sigma factor, whose expression is regulated by ComE, does not participate in this signaling pathway [34]. We have also reported that ComDE and CiaRH control pneumococcal survival in pneumocytes in contrasting ways, CiaRH was essential for ATR and intracellular survival, whereas ComE repressed its activation. Moreover, ComE in a CSP-independent manner, was necessary for ASIL, whereas CiaRH protected against its induction by modulating LytA autolysin expression on the pneumococcal surface. These results suggest that both TCSs control the acidic stress response and establish either a survival or a suicidal response by independent pathways, either in acidified culture media or in pneumocyte cultures [33]. These findings indicate that ComE is activated under acidic conditions by an alternative signaling pathway that differs from the quorum sensing mechanism reported during competence development at alkaline pH. Alternatively, it was proposed that StkP is involved in competence at pH 7.8, by the fact that cells lacking StkP do not develop natural competence [4,7,22]. StkP is also essential for virulence, to establish infections in the lung and for invading and growing in the bloodstream of intranasally infected mice [4].

The main aim of this work was to elucidate whether ComE is part of a novel activation pathway used by S. pneumoniae to induce the acidic stress response and to control its intracellular survival mechanism in pneumocytes. Here, we demonstrate that StkP controls ComE activation by phosphorylation of the Thr128 residue of the latter, increasing both its dimerization capacity and its DNA-binding affinity. Under acidic conditions, the StkP/ComE HK-independent pathway regulated 104 genes involved in different cellular processes, such as H2O2 production and oxidative stress tolerance. The StkP/ComE pathway is independent of the HK-dependent ComD/ComE system, which regulates more than 180 genes at pH 7.8 [35]. The participation in HK-independent and HK-dependent stress regulatory systems places ComE as a global regulator. This newly discovered StkP/ComE signaling pathway triggered the acidic stress response by inducing ASIL and inhibiting ATR and the intracellular survival of S. pneumoniae in pneumocytes, one of the first barriers that this pathogen must overcome to establish an infection.

Results

The ComE response regulator is activated under acidic conditions by a histidine-kinase independent pathway

We previously reported that ComE was required in the acidic stress induced lysis (ASIL) mechanism, which was independent of its cognate histidine kinase ComD at pH 6.0 [34]. This is in contrast to the ComD/quorum-sensing dependence on ComE activation at pH 7.8 necessary for competence development [19]. This initial observation led us to investigate whether other pneumococcal TCS-associated HKs could be activating ComE by a crosstalk mechanism, as described for other bacteria [36]. We hypothesized that if other HKs were involved in ComE activation by phosphorylation, the corresponding hk mutant should display alterations in the ASIL induction. Thus, the lytic phenotype was determined under acidic stress conditions for all the pneumococcal hk mutants that we had previously constructed in the background of the R801 strain by insertion-duplication mutagenesis (S1 Table) [33]. We observed that all the hk mutants showed the same lytic response as the parental R801strain, indicating that none of the tested HKs was involved in ASIL (S2 Table). In addition, the comDF183X mutant was used because it lacks the HK domain due to a stop codon at residue 163 (S2 Table) and therefore it is unable to activate ComE [33]. This mutant was constructed to avoid putative alterations in the comCDE operon expression, and it showed the same ASIL phenotype than the ΔcomD mutant (S2 Table), indicating that the truncated ComD protein expressed by the comDF183X mutant has not impact on the ASIL activation. In order to avoid a putative residual effect of comDF183X on the ComE activation, we determined ASIL in the double comDF183X hk mutants, which were constructed by transforming the comDF183X mutant with individual plasmids containing the different hk mutations. Despite the low transformability of the comDF183X mutant, all the double comDF183X hk mutants displayed the same ASIL phenotype as those obtained for each individual hk mutant (S2 Table). Taken together, these results indicated that HKs were not responsible for activation of ComE and the resulting ASIL.

Although ASIL is controlled by ComE activation without HK participation, this finding does not exclude the potential that ComE could have been phosphorylated by another phosphodonor at the Asp58 residue, typically the target of ComD phosphorylation [18]. Prokaryotic response regulators can be phosphorylated in vivo by acetyl-phosphate at the conserved aspartate residue of the receiver domain, resulting in similar activation to that exerted by the cognate HK [37]. In addition, phosphorylation crosstalk between HK and RR that belong to different TCSs has been reported [38]. Therefore, we first analyzed the possibility that Asp58 phosphorylation could be required for ASIL, and tested the comED58A mutant that encodes for the ComED58A protein, in which the phosphorylatable Asp58 residue is replaced by alanine [18,33]. The presence of this mutation was phenotypically corroborated under competence development conditions, confirming that CSP-induced competence was eliminated in the comED58A mutant (S1 Fig and [18]). Like the wt R801 strain, the comED58A mutant autolysed under acidic conditions (Fig 1A) indicating that Asp58 phosphorylation is not necessary for ASIL. The comED58A phenotype is similar to the phenotype displayed by the hk mutants. Taken together, these results suggest ComE activation under acidic conditions is independent of both Asp58 phosphorylation and HK activity.

thumbnail
Fig 1. Evaluation of ASIL and comE expression in S. pneumoniae mutants.

Autolysis was measured as a change in OD620nm over 6 hours. Lytic curves corresponding to specific mutants are indicated in each panel (A-C), with data being representative of at least three independent experiments. (A) ASIL is controlled by StkP but it does not require Asp58-phosphorylation in ComE. (B) StkP does not participate in the CiaRH-regulated ASIL pathway. (C) StkP is involved in the ComE-regulated ASIL pathway. References: *p< 0.05; **p< 0.01; ***p< 0.001, these p-values were referred to the wt strain in each panels. (D) Transcription levels of the comE gene measured in cells exposed to pH 6.0. To avoid autolysis, all mutants were constructed in a ΔlytA (autolysin deficient) background. The ΔlytA, comED58A ΔlytA, ΔstkP ΔlytA, comDT233I ΔlytA and comET128A ΔlytA cells were grown in ABM/pH 7.8 to the mid-exponential phase and resuspended in ABM/pH 6.0. Total RNA was extracted at 0 min, 10 min, and 30 min. The fold change in gene expression was measured by quantitative real-time PCR and calculated using the 2–ΔΔCT method. The gyrB gene was used as the internal control and the reference condition was time 0 min of strain ΔlytA. Error bars indicate the standard deviation of the mean. INSTAT software was used to perform Dunnet’s statistical comparison test for each strain with its respective basal condition (time 0 min). References: **p< 0.01; ***p< 0.001.

https://doi.org/10.1371/journal.ppat.1007118.g001

The serine/threonine kinase StkP is essential for ASIL activation and participates in the ComE pathway

Since it has been shown that StkP is involved in comCDE expression during competence development at pH 7.8 [4], and that ComE is indispensable for the induction of ASIL [34], we evaluated whether StkP participated in ASIL development. Thus, the ΔstkP mutant strain did not autolyze when cultured under acidic conditions (Fig 1A), strongly suggesting that StkP is necessary for ASIL induction. To further confirm whether StkP kinase activity was required for ASIL, we also constructed the stkPK42R mutant, which encodes for StkP with reduced enzymatic activity, as previously described [39]. This reduced kinase activity produces multiple septa, peripheral peptidoglycan biosynthesis and elongated cells [8], and these alterations were confirmed in our mutant and compared with the wt strain (S2A and S2B Fig). As expected, the stkPK42R mutant strain showed ASIL with a degree of autolysis inferior to the wt strains but higher than the ΔstkP strain, indicating that the residual kinase activity in the stkPK42R mutant [39] was likely responsible for ASIL induction. These observations strongly suggest that StkP kinase activity is essential for ASIL activation.

To exclude the possibility that the ASIL blockage observed in the ΔstkP mutant is a side effect due to hampered cell division and/or compromised cell wall structure [13], we constructed another mutant that presents cell division alterations, such as the ΔmapZ mutant [9]. We verified these alterations by Van-Fl staining (S2C Fig) and found that the ΔmapZ mutant showed an ASIL phenotype similar to the wt strain (S2D Fig), indicating that the ASIL effect showed by the ΔstkP mutant is independent of cell division alterations.

We have previously demonstrated that ASIL is controlled by two independent signaling pathways, CiaRH and ComE. While CiaRH plays a protective function, the ComE acts by promoting ASIL [34]. To determine whether StkP participated in the CiaRH-controlled ASIL pathway, the ΔstkP and ΔciaR mutations were backcrossed, and the lytic phenotype of the resulting double mutants was analyzed. The ΔstkP ΔciaR strain showed enhanced autolysis, similar to the ΔciaR single mutant, demonstrating that the ciaR mutation had an epistatic effect on the non-autolytic stkP phenotype and that StkP activity did not participate in the signaling events of the CiaRH-controlled ASIL pathway (Fig 1B).

It has been previously described that the comDT233I mutation produces a hyperactive ComD HK, which in turn hyperphosphorylates ComE, activates comCDE transcription, and results in high intracellular levels of ComE [40]. Although our results suggested that ComD was not involved in ComE-mediated ASIL, we constructed the comDT233I mutant to artificially produce high levels of ComE and competence, as described [34,40]. The comDT233I mutant showed constitutive high levels of comE transcripts, at 1000-fold higher compared to the wt strain. This mutant displayed accelerated ASIL compared to the wt strain, however, we previously reported that ASIL was blocked in the double comDT233I ΔcomE double mutant [34]. These results demonstrated that ASIL induction was ComE-dependent and that increase expression of the latter leads to accelerated autolysis [34]. More importantly, when the stkP gene was disrupted in the comDT233I mutant, ASIL was also completely blocked (Fig 1C), suggesting that StkP is essential for ASIL activation despite the high ComE levels expressed in the comDT233I mutant.

comE expression is induced in response to acidic conditions

It is known that comE is part of the comCDE operon, with the transcription of this operon initiated at the pcomC promoter (bp 2035421–2035806, S. pneumoniae R6 genome, NCBI reference: NC_003098.1) during competence development at alkaline pH [41]. To test whether pcomC was responsible for the increase in comE observed under acidic conditions, we constructed the pcomC-lacZ reporter fusion (S1 Table), which was integrated via a single crossover event upstream of comC in a bgaA mutant (deficient in β-galactosidase activity). When the bgaA mutant strain carrying the pcomC-lacZ fusion was incubated at pH 6.0 for 30 min., a 1.7-fold increase in β-galactosidase activity was observed. No such increase was detected in a ΔcomE knocked out mutant (S3 Fig). To further confirm this observation, the levels of comE transcript in the wt strain were determined by qPCR, which showed a 4-fold increase in cells exposed for 30 min at pH 6.0 (Fig 1D). These results indicate that the increased number of comE transcripts was caused by acidic stress, with this activation being dependent on ComE. Similarly, the levels of comE transcript in the comED58A mutant were increased 4.5-fold after 30 min incubation at pH 6.0. This result indicates that ComED58A was able to induce ASIL under acidic stress conditions (Fig 1D). Consequently, these results suggest that ComE is activated by an alternative signaling pathway that does not require phosphorylation of Asp58.

We previously reported that induction of comE transcripts by acidic stress is a characteristic of the ComE-mediated pathway that controls ASIL [34]. To examine whether StkP could be involved in this pathway, we analyzed the comE transcript levels by qPCR in the ΔstkP and stkPK42R mutants constructed in a lytA background to avoid autolysis (S1 Table). After incubation of ΔstkP cells for 30 min at acidic pH 6.0, we observed a five-fold reduction in the levels of comE transcripts in the ΔstkP mutant (Fig 1D). In contrast, the stkPK42R mutant showed a 2-fold decrease in comE transcripts, likely due to reduced kinase activity of StkPK42R. In addition, we observed that StkP was capable of controlling pcomC activation by acidic stress since increased β-galactosidase activity was observed from the pcomC-lacZ reporter fusion presence in the ΔstkP mutant in the bgaA background (S3 Fig). Taken together, these results indicate that StkP kinase activity was required for comE induction under acidic conditions.

In the comDT233I ΔstkP double mutant, the levels of comE transcript increased 50 times over those in the ΔstkP strain and 10 times over those in the wt strain (Fig 1D). These results suggest that StkP kinase activity is required for full activation of ComE in order to induce ASIL under acidic conditions, regardless of the presence of high levels of ComE unnaturally induced by the ComDT233I kinase. Such observations led us to speculate that StkP activates ComE by an alternative mechanism other than the classical ComD HK-mediated Asp58 phosphorylation.

ComE is phosphorylated in vitro and in vivo by StkP

We hypothesized that StkP controls ComE by a crosstalk phosphorylation event. To test this hypothesis, we carried out an in vitro phosphorylation assay using purified recombinant Hisx6-ComE fusion protein, in the presence or absence of purified recombinant GST-StkP. The phosphorylation reactions were examined by immunoblotting using either anti-phosphoserine or anti-phospho-threonine antibodies. No signal was detected with the anti-phosphoserine antibody, in contrast, positive reactions were detected with the anti-phosphothreonine antibody (Fig 2A), with the phosphorylation reaction occurring at a molar ratio range of GST-StkP/Hisx6-ComE between 1:2 and 1:20 (Fig 2B). The GST-GFP or Hisx6-GFP were included as controls of reaction specificity, and Hisx6-DivIVA was used as a positive control of a StkP target, as previously described [8,11] (Fig 2A). We also evaluated the possibility that StkP could trigger ASIL by phosphorylating the major pneumococcal autolysin LytA. As overexpression of the full-length LytA protein was toxic in E. coli cells, the N-terminal region of LytA, which contains the catalytic domain was expressed instead fused to a Hisx6-tag (N-LytA-Hisx6)[42]). Incubation with or without GST-StkP, resulted in no evidence of N-LytA-Hisx6 phosphorylation (Fig 2A), suggesting that LytA was not phosphorylated by StkP, at least under the experimental conditions described here.

thumbnail
Fig 2. ComE is phosphorylated by StkP.

(A) ComE is phosphorylated at a threonine residue by StkP. Top: nitrocellulose membrane stained with Ponceau S as a loading control. Bottom: Immunodetection of phosphorylated proteins. Phosphorylation reactions were carried out with purified GST-StkP and substrate proteins (0.5 μg each) mixed in kinase buffer and incubated at 37°C for 1 hour. Phosphorylated proteins were detected with an anti-phosphothreonine polyclonal antibody. Lane 1: Hisx6-ComE. Lane 2: Hisx6-ComE + GST-StkP. Lane 3: Hisx6-GFP. Lane 4: Hisx6-GFP + GST-StkP. Lane 5: LytA(N)-Hisx6. Lane 6: LytA(N)-Hisx6 + GST-StkP. Lane 7: Hisx6-DivIVA. Lane 8: Hisx6-DivIVA + GST-StkP. (B) ComE phosphorylation assays with different StkP:ComE molar ratios. GST-StkP and Hisx6-ComE were mixed at different molar ratios in kinase buffer and incubated at 37°C for 1 hour. Detection of phosphorylated proteins was performed as described above. (C) In vivo StkP-dependent and acid-induced ComE phosphorylation. C-terminal His-tagged ComE was purified from wt and ΔstkP strains grown in ABM (pH 7.8), and exposed to acidic stress in medium MD5, pH 6.0. Protein samples were separated by SDS-PAGE and phosphorylated or total ComE-His was detected with Pro-Q Diamond and SYPRO Ruby staining, respectively.

https://doi.org/10.1371/journal.ppat.1007118.g002

To determine whether StkP-mediated ComE phosphorylation occurs in vivo and because response regulators are usually expressed at low level in bacteria, we constructed by insertion-duplication mutagenesis wt and ΔstkP strain derivatives that express ComE fused to the Hisx6-epitope tag at the C-terminus (ComE-His6x) to improve ComE detection. Cells were incubated at either pH 7.8 or pH 6.0 and ComE-Hisx6 was purified from protein lysates as described in Material and Methods and separated by SDS-PAGE. Phosphoproteins were detected by ProQ Diamond staining while total proteins were detected by SYPRO Ruby staining. We observed that phosphorylated form of ComE in wt cells grown at pH 7.8, that increases 2.3 times when cells are grown at pH 6.0 (Fig 2C). In contrast, ComE remained unphosphorylated in the ΔstkP mutant, confirming that ComE is phosphorylated by StkP in vivo.

StkP phosphorylates ComE at the Thr128 residue to regulate ASIL

To determine the amino acid residues in ComE that are targeted for phosphorylation by StkP, we performed HPLC-MS/MS analysis of the in vitro StkP-phosphorylated ComE-His6x recombinant protein. A single amino acid was identified as a target for StkP-mediated phosphorylation in His6x-ComE, Thr128, located inside the trypsin-digested 121IEQNIFYTK129 ComE peptide (Fig 3A). To further confirm this observation, we created the non-phosphorylatable ComET128A-His6x recombinant mutant protein that remained unphosphorylated in the presence StkP in vitro (Fig 3B). To evaluate the role of Thr128 phosphorylation on ComE activity in vivo, we constructed the comET128A mutant, which showed significantly blocked autolysis compared to the wt (Fig 3C). Using the comET128A mutant, we produced the revertant comEA128T strain, which showed an ASIL phenotype similar to the wt strain (Fig 3C). To further support the role of ComE Thr128 phosphorylation in ASIL activation, we attempted to replace Thr128 by Glu128 to construct the phosphomimetic comET128E protein, which is typically used to mimic the phosphorylated form of Thr residues [43]. In vitro, the phosphomimetic ComET128E-Hisx6 protein was hyper-activated, as demonstrated by EMSA assays (see next), which may explain our inability to produce a viable comET128E mutant. These assays confirmed that Thr128 phosphorylation is essential for the StkP-mediated ComE activation that controls ASIL and that ComE hyper-activation is likely lethal to S. pneumoniae.

thumbnail
Fig 3. StkP phosphorylates ComE on the Thr128 residue to control ASIL.

(A) To identify the phosphorylation site, tryptic peptides obtained from ComE previously incubated with StkP were analyzed by nano-LC-MS/MS. The figure shows the MS/MS spectrum of the di-charged ion of m/z 618.8 corresponding to the phosphorylated sequence IEQNIFYTK. C-terminal y ions are labeled in blue, while N-terminal a or b fragment ions are labeled in red. Ions containing pT residue present the phosphorylation characteristic neutral loss of 98 Da. Thr128 is unequivocally identified as the phosphorylated residue (Xcorr 3, 45; pRS score 148). (B) StkP phosphorylates ComE at Thr128. In vitro phosphorylation assays were performed with purified GST-StkP and Hisx6-ComEwt or Hisx6-ComET128A proteins mixed in kinase buffer at a StkP/ComE ratio of 1:20. Phosphorylated proteins were detected with an anti-phospho-threonine polyclonal antibody. Lane 1: Hisx6-ComE. Lane 2: Hisx6-ComE + GST-StkP. Lane 3: Hisx6-ComET128A. Lane 4: Hisx6-ComET128A + GST-StkP. (C) ASIL requires the Thr128 residue in ComE for lysis induction. Autolysis was determined as indicated in the legend of Fig 1. Lytic curves corresponding to specific mutants are indicated, which data is representative of at least three independent experiments. References: ***p> 0.001.

https://doi.org/10.1371/journal.ppat.1007118.g003

Competence regulation is independent of Thr128 phosphorylation in ComE

StkP is involved in competence in response to stress conditions such as pH 7.8 [2,4,7]. Since, our results indicated that a signaling pathway that involves StkP and ComE controls autolysis in response to acidic stress at pH 6, we investigated whether this crosstalk mechanism could also regulate competence development at pH 7.8. We have previously described that the stkP mutant showed no competence development at pH 7.8 [4]. In contrast, the comDT233I mutant shows a hypercompetent phenotype, accompanied by constitutively high levels of ComE expression [34]. We observed that the competence phenotype of the comDT233I ΔstkP double mutant was similar to the comDT233I single mutant (S1 Fig), indicating an epistatic effect of the comDT233I mutation on the ΔstkP mutation. We also observed that the competence of the non-phosphorylatable comET128A and revertant comEA128T mutants were similar to the wt strain (S1 Fig).

These observations suggest that StkP regulates competence at an early stage, which is independent of ComE Thr128 phosphorylation. Furthermore, StkP was not essential for competence once ComE was activated by ComD at pH 7.8. In contrast, StkP is necessary to activate ComE and to trigger autolysis under acidic conditions.

StkP-mediated Thr128 phosphorylation increases ComE dimerization

Response regulators are composed by a conserved receiver domain, which is phosphorylated on an aspartate residue by their cognate histidine kinase, and DNA-binding domains [44]. In ComE, the receiver domain corresponds to the first 130 residues [43]. Thr128 is located at the end of the α-5 region (Asp114-Ser130) of the receiver domain of ComE, next to the α-4 region (Ala94-Gln101), and near the loop between α-4 and β-5 (Val102-Leu105) that is involved in ComE dimerization and considered as a dimerization interface [43] (Fig 4A). The proximity between the dimerization interface and the two phosphorylated residues (Asp58 and Thr128) suggest a putative influence on the dimerization capacity of ComE, which was confirmed by in vitro dimerization assays using the phosphomimetic mutants. The ComED58E-His6x, ComET128E-His6x, and ComET128A-His6x mutants showed dimer steady-state levels, which were 70, 72 and 2.9 times higher, respectively, than ComEwt-His6x (reference level). In addition, when the ComEwt-His6x protein was incubated with StkP, the dimerization rate increased 8.9 times, whereas ComET128A-His6x showed only a 3.3-fold increase (Fig 4B). The different dimerization capabilities found for ComEwt/StkP (8.9 times) compared to the phosphomimetic ComET128E-His6x (70 times) suggest that ComEwt-His6x is partially phosphorylated by StkP (~12%). These observations suggest that Thr128 phosphorylation modifies ComE in a manner that strongly affects its dimerization interface, which is a condition sine qua non for the response regulator activation.

thumbnail
Fig 4. Thr128-phosphorylation increases the dimeric state of ComE.

(A) Localization of Thr128 residue in the ComE structure. Based on the crystal structure of ComE reported by Boudes et al [43], this figure reveals the localization of the Thr128 residue, as well as the alternative phosphorylation site Asp58. The three loops in the DNA-binding domain are also shown, which are apparently altered when ComE is phosphorylated on Thr128. At the bottom of this image, a sequence alignment between the DNA-binding domains of ComE and AgrA is also shown. Positively charged or polar residues, which are described in AgrA to have a direct contact with DNA bases [45], are indicated in red. (B) The dimerization capacity of recombinant ComE proteins, such as the phosphomimetic ComED58E and ComET128E proteins, as well as the non-phosphorylatable ComET128A mutant, was analyzed and compared with ComEwt (left panel). ComEwt and ComET128A were also pre-incubated with GST-StkP (right panel). Dimerization states were assessed by native PAGE/Tris-MOPS buffer. Proteins were electroblotted onto PVDF membranes, and Hisx6-ComE was detected using anti-His antibody.

https://doi.org/10.1371/journal.ppat.1007118.g004

Thr128 phosphorylation changes conformation of the ComE DNA-binding domain

In order to determine how Thr128 phosphorylation affected ComE's conformation, we performed molecular dynamic simulations at 40–150 ns comparing ComEwt (PDB ID: 4CBV, [43]) and the in silico phosphomimetic ComET128E-His6x protein (S4 Fig, video). The simulations clearly indicated that 3 loops spanning the DNA-binding domain (Lys218-Asn219-Leu220, Thr164-Gly165-Val166-Ser167-His168, and Ser200-Pro201-His202-Lys203) presented different dynamics in the ComET128 mutant compared to ComEwt (Fig 4B and accompanying video shown in S4 Fig). ComE is a member of the AgrA/LytTR family of bacterial response regulators, which present certain structural homologies. Coincidently, two of these three loops have been described for the AgrA RR in S. aureus [45], as key residues that determine the DNA binding affinity for promoters in the phosphorylated form of AgrA (Fig 4B). ComE also revealed positively charged or polar residues (His168 and Lys169 in loop 1; His202 and Lys203 in loop 2; Arg217 and Lys218 in loop 3), which are shown in AgrA to have a direct contact with DNA bases [45]. These results suggest that after Thr128 phosphorylation ComE may undergo conformational changes. Consistent with this notion, limited proteolysis assays revealed structural differences between ComE-His6x and ComET128E-His6x (S5 Fig). Treatment with trypsin showed more contrasting proteolytic patterns than proteinase K treatment. These findings confirmed that Thr128 phosphorylation causes evident changes in ComE conformation, likely in the DNA-binding domain of ComE, that may modify its DNA-binding affinity, and we explored this possibility with electrophoretic mobility shift assays (EMSAs).

StkP-mediated Thr128 phosphorylation in ComE increases its DNA-binding affinity in a pH-dependent manner

During competence development at alkaline pH, Asp58 phosphorylation by ComD results in increased binding of ComE to pcomC and transcriptional activation of the comCDE operon [2,46,47]. We have observed that at acidic pH, pcomC activation and induction of comE transcription depended both on ComE and on StkP (Fig 1D and S3 Fig) suggesting that Thr128 phosphorylation by StkP influences the binding of ComE to pcomC. Electrophoretic mobility shift assays (EMSAs) proved that the phosphomimetic ComET128E-His6x protein bound pcomC 5-fold stronger than ComEwt-His6x (Kd 74 nM vs Kd 371 nM, respectively). The DNA binding affinity of the non-phosphorylatable ComET128A-His6x mutant was unaffected (Kd 375 nM) whereas ComED58E-His6x affinity for pcomC was 17- fold greater that ComEwt-His6x (Fig 5 and Table 1). Curiously, when ComEwt-His6x was pre-incubated with StkP in phosphorylation buffer at pH 7.8 no binding to pcomC was observed (S6 Fig, Table 1). As ASIL is regulated by StkP-mediated phosphorylation of ComE Thr128 residue under acidic conditions, we tested if ComE DNA-binding affinity could be affected by pH. When ComEwt-His6x was previously incubated with StkP at pH 6.0, its affinity for pcomC increased (Kd 64 nM) and was similar to that shown by ComET128E-His6x (S6 Fig, Table 1). To determine which of these contrasting effects actually depended on StkP phosphorylation, similar assays were performed with an inactive StkP enzyme (StkPK42M) [8] and ComEwt-His6x.

thumbnail
Fig 5. The phosphomimetic ComET128E protein shows an increased DNA-binding affinity.

The DNA-binding affinity for the promoter region of the comCDE operon (pcomC) of ComEwt (A), the non-phosphorylatable (by StkP) ComET128A mutant (B) and the phosphomimetic ComET128E (C) and ComED58E (D) proteins was determined by EMSA. Binding interactions were examined by incubating variable amounts of the different ComE versions with Cy5-labeled pcomC, followed by electrophoretic separation of the protein-DNA complexes. Black or white triangles are indicating the free or ComE-bound probe, respectively. Images were obtained with a fluorescence scanner as described in Materials and Methods. The Kd values are indicated in each panel.

https://doi.org/10.1371/journal.ppat.1007118.g005

Binding to pcomC was comparable in ComEwt-His6x pre-treated with StkPK42M at pH 6.0 and untreated ComEwt (Kd 300 nM vs Kd 371 nM) but was still absent after incubation at pH 7.8 (S7 Fig, Table 1). These results indicate that StkP phosphorylation at pH 6.0 underlied the enhanced pcomC-binding affinity of ComE, but not the blocking of ComE-pcomC interaction observed at pH 7.8, which suggests that at a slightly alkaline pH, StkP makes a complex with ComE masking its DNA-binding sites. The DNA-binding affinity of the phosphomimetic ComET128E-His6x and the non-phosphorylatable ComET128A-His6x proteins were not affected when preincubated with StkP at either pH 6.0 or pH 7.8 (S7 Fig, Table 1), indicating that Thr128 mediated the observed StkP effects on ComE: (1) at pH 6.0, Thr128 phosphorylation by StkP increases ComE DNA binding affinity; (2) at pH 7.8 Thr128 mediates the StkP-ComE interaction that blocks DNA binding. Thus, these experiments indicate that pH modulates the interplay between StkP and ComE.

The StkP/ComE interaction increases at acidic pH

To test for a putative protein-protein interaction between StkP and ComE, a sandwich fluorescence-linked immunosorbent assay (FLISA) was utilized, in which the StkP-coated surface of a microtiter plate was incubated with increasing amounts of His-tagged ComE at either pH 6.0 or pH 7.8. This assay clearly showed that acidic pH augmented the number of binding sites between ComE and StkP, as reflected by a 3-fold increase in Fmax (maximum fluorescence when binding is saturated) at pH 6.0 compared to pH 7.8 (Table 2, S8 Fig). The interaction between ComEwt and the StkPK42M mutant revealed the same Fmax as the ComE/StkP at different pH values, indicating that kinase activity did not alter the saturation of this protein complex. However, for the StkPK42M mutant, the K1/2 values were lower, indicating that StkPK42M bound more tightly to ComE at any pH value. In comparison with ComEwt, the interaction between non-phosphorylatable ComET128A mutant and StkP or StkPK42M was 3-fold stronger and was not affected by pH. In contrast, the phosphomimetic ComET128E protein produced a 3-fold increment in K1/2 at pH 7.8, which further raised to 9-fold at pH 6.0, demonstrating that the phosphorylated form of ComE had a lower affinity for StkP (Table 2, S8 Fig).

thumbnail
Table 2. Estimates of ComE binding affinities for StkP and StkPK42M.

https://doi.org/10.1371/journal.ppat.1007118.t002

These data confirm that the StkP/ComE interactions are mediated by ComE residue Thr128 and favored by acidic conditions, which may facilitate ComE phosphorylation.

ComE induces global changes in the transcriptome of S. pneumoniae

To understand the effect of the StkP/ComE signaling pathway on pneumococcal physiology, we compared the transcriptomes of the comET128A mutant and wt by RNAseq analysis. Three replicates of each strain strains were grown in ABM (pH 6.0) for 1 hr at the exponential growth phase (OD620nm 0.3) and analyzed. In total, the differential expression of 104 genes was detected, 51 were down-regulated genes and 53 were up-regulated, considering relevant genes to be those with expressions higher than 2 fold and p values <0.05 (Fig 6A). The full list of these genes is shown (S3 Table). Based on this differential gene expression analysis, we observed that the StkP/ComE pathway affected, directly or indirectly, the expression of genes involved in oxidative stress, and the purine/pyrimidine, amino-acid and central metabolisms, as well as the ribosomal and translation structures, metabolite transport, molecular chaperones and cell wall biosynthesis, among others (Fig 6B). The list of genes regulated by Thr128-phosphorylated ComE indicated that this new signaling pathway induces global changes in the pneumococcal transcriptome, such as the physiological response to acidic stress. Bioinformatic analysis of the promoter regions (240 bp upstream of the start codon) of 22 ComE-regulated genes obtained from RNAseq assays predicted a putative DNA binding motif (S9A Fig). Martin et al [47] described a potential ComED58~P binding site (CEbs, 32 bp) in the comC promoter constituted by two repeats (DR1 and DR2) separated by 12 bp. In this report, we established that the putative ComET128~P binding site (26 bp) only partially overlaps with theses repeats suggesting a different consensus binding sequence to that described for ComED58~P (S9B Fig).

thumbnail
Fig 6. ComE is a global regulator that controls gene expression during the stress response.

(A) Gene expression scatter plot in the wt and comET128A samples, with the x-axis representing the gene expression values for the control condition (wt) and the y-axis representing those for the treated condition (comET128A). Each black dot represents a significant single transcript, with the vertical position of each gene representing its expression level in the experimental conditions and the horizontal one representing its control strength. Thus, genes that fall above the diagonal are over-expressed whereas genes that fall below the diagonal are underexpressed as compared to their median expression levels in the experimental groups. (B) Volcano plot of gene expression in wt vs comET128A samples measured by RNAseq. The y-axis represents the mean expression value of the log10 (p-value), while the x-axis displays the log2 fold change value. Black dots represent genes with an expression 2-fold higher in the comET128A mutant relative to strain wt with a p-value < 0.05, with red dots signifying genes with an expression 2-fold lower in the comET128A mutant, which are relative to strain wt with a p < 0.05. (C) Categories of ComE-regulated genes obtained from an RNAseq analysis. An RNAseq generated distribution in functional categories of genes that are regulated in the comET128A mutant relative to strain wt under acidic conditions. (D) ComE-regulated genes expressed under acidic conditions in the comET128A mutant relative to strain wt. Gene expression determined by RNAseq was confirmed by qPCR. The comET128A ΔlytA and ΔlytA (referred as wt for this assay) strains were grown in ABM/pH 6.0 to the mid-exponential phase in triplicate, with the fold change in gene expression measured by RT-qPCR and calculated using the 2–ΔΔCT method. The gyrA gene was used as the internal control. References: ** p < 0.01; *** p < 0.001.

https://doi.org/10.1371/journal.ppat.1007118.g006

The StkP/ComE pathway controls H2O2 production and oxidative stress response

Using RT-qPCR we confirmed decreased expression of oxidative stress genes spxB, sodA [48] and tpxD [49] in comET128A mutant compared to wt (Fig 6C). The spxB gene encodes the pyruvate oxidase that produces H2O2 from O2, sodA encodes the superoxide dismutase that produces H2O2 from superoxide, and tpxD encodes the thiol peroxidase that catalyzes the H2O2 oxidation and contributes to the oxidative stress response. In the comET128A mutant, we found that the spxB, sodA, and tpxD transcripts were downregulated 18, 2.8 and 2.7 times, respectively (Fig 6C). These findings were also corroborated by H2O2 production and H2O2 susceptibility assays. The comET128A, ΔcomE, and ΔstkP mutants showed a 4-fold decrease in their H2O2 production compared to the wt and comEA128Tstrains (Fig 7A), which is likely caused by the reduced expression of spxB and sodA protein products. In addition, we observed a 10-fold reduction in the susceptibility to H2O2 by the comET128A, ΔcomE, and ΔstkP mutants compared to the wt strain (Fig 7B), likely due to reduced expression of the TpxD peroxidase. These findings support the notion that the StkP/ComE pathway is essential for the control of H2O2 production and for H2O2 tolerance.

thumbnail
Fig 7. The StkP/ComE pathway controls oxidative stress and cell wall biosynthesis.

(A) The H2O2 production is altered in the comE and stkP mutants. Cells were grown in BHI at 37°C to an OD620nm of 0.3, then diluted in either ABM (pH 6.0) and incubated at 37°C to an OD620nm of 0.3. The H2O2 concentration was determined by the peroxidase test as described in Material & Methods. Values were calculated as the H2O2 concentration in mM and normalized against the number of viable cells. (B) The comE and stkP mutants were more susceptible to H2O2 than wt. Susceptibility to H2O2 is indicated as a percentage of bacterial survival at different time points. C-D) The comET128A mutant was more resistant to cell-wall antibiotic-induced lysis than wt. Cells were grown in BHI/pH 7.2 at 37°C to an OD620nm of 0.3, and fosfomycin (C) and vancomycin (D) were added in independent cultures at final concentrations of 50 μg/ml and 0.4 μg/ml, respectively. Cell lysis of bacterial cultures was determined by turbidimetry at OD620nm for more than 3 h. References: *** p < 0.001.

https://doi.org/10.1371/journal.ppat.1007118.g007

The StkP/ComE pathway regulates murN expression and modulates susceptibility to antibiotic-induced lysis

Although RNAseq analysis showed that the murN gene was overexpressed in the comET128A mutant, its expression by qPCR was actually found to be 4-fold lower than in wt in three independent assays (Fig 6C), suggesting a typical case of false positive that is commonly found in RNAseq studies. Regarding the physiological impact of the murN mutation, Filipe et al [50] described that a murMN mutant had cell wall alterations and presented increased susceptibility to lysis when exposed to cell wall antibiotics. To test whether an altered murN expression in the comET128A mutant could modify the susceptibility to cell wall antibiotics, we determined the MIC values of the comET128A and wt strains in the presence of either fosfomycin, vancomycin, penicillin, cefotaxime, cefazolin, or piperacillin. The fosfomycin MIC in the comET128A (170 μg/ml) was higher than the wt strain (50 μg/ml), whereas the MICs for vancomycin, penicillin, cefotaxime, cefazolin, and piperacillin were similar between these strains. The typical lytic effect of fosfomycin (50 μg/ml, 1xMIC; Fig 7C) and vancomycin (0.4 μg/ml, 1xMIC; Fig 7D) on the wt strain was inhibited in the comET128A strain. The diminished susceptibility to cell wall antibiotics in the comET128A strain suggests cell wall alterations consistent with the ASIL repression showed by this mutant.

Pneumococcal survival in pneumocytes is controlled by the StkP/ComE pathway

We previously described that ComE is involved in the acidic stress response and in the pneumococcal intracellular survival mechanism in pneumocytes [33]. Here, we demonstrate that StkP phosphorylates ComE, and in order to determine whether this crosstalk affects the pneumococcal survival, we measured the intracellular survival capacities in A549 pneumocyte cells of the ΔstkP, stkPK42R (reduced kinase activity), ΔcomE, comET128A and comEA128T (revertant) strains compared to the wt in A549 pneumocyte cells [33]. Mutations in either the stkP or comE genes conferred increased survival compared to comEA128T or wt (Fig 8A), indicating that the StkP/ComE pathway controlled pneumococcal survival in pneumocytes. To discriminate whether this phenotype could result from increased ATR or decreased ASIL, we tested the ΔlytA mutant, which lacks autolysin and presented a blocked ASIL [33], but its intracellular survival was similar to the wt strain (Fig 8A). Thus, a blocked ASIL is not enough to increase intracellular survival of S. pneumoniae in pneumocytes. Consequently, the increased survival showed by either the ΔstkP, ΔcomE, or comET128A mutants (Fig 8A) is likely due to higher ATR capacity. To test this hypothesis, we determined the ATR phenotype of the comET128A mutant, but in a lytA deficient background in order to discard residual autolysis. As expected, ATR of the ΔlytA strain increased 2-fold at pH 6.0 compared with cells cultured at pH 7.8, whereas the comET128A ΔlytA cells showed a 20-fold increase under the same conditions (Fig 8B), supporting the notion that increased ATR explains the increased survival rate displayed by the comET128A mutant in pneumocytes.

thumbnail
Fig 8. The StkP/ComE pathway modulates intracellular survival and the acid tolerance response of S. pneumoniae.

(A) The ΔstkP and ΔcomE mutants showed increased intracellular survival compared with wt in A549 pneumocytes. Bacteria cells were initially incubated for 3 h in monolayers of A549 pneumocytes, and survival progression of different strains was monitored using a typical protection assay. Survival percentages were calculated by considering the total amount of internalized bacteria after 30 min of extracellular antibiotic treatment as representing 100% for each strain. After antibiotic treatment, samples were taken at 0 (white bars), 3 (grey bars) and 7 (black bars) hours, and pneumocytes were lysed to release pneumococci. Samples were diluted in BHI spread on BHI-blood-agar plates and incubated at 37°C for 16 h. (B) The ΔstkP and ΔcomE mutants displayed an augmented ATR compared with wt. To determine the survival percentage of bacterial strains, the non-induced cells (white bars) were directly exposed for 2 h at pH 4.4 (lethal pH) in THYE medium, with the acid-induced cells (grey bars) being previously incubated for 2 h at pH 6.0 (sub-lethal pH) in THYE medium. After exposition to lethal pH, pneumococcal survival was determined by spreading dilutions in BHI-blood-agar plates and incubating these at 37°C for 16 h. For both panels, data are representative of at least three independent experiments and statistically significant differences are indicated as p<0.01 (**) or p<0.001 (***).

https://doi.org/10.1371/journal.ppat.1007118.g008

Discussion

Two-component systems (TCSs) represent one of the most important mechanisms of gene regulation in bacteria. Alternatively, eukaryotic-like serine-threonine kinases (STKs) constitute another signaling mechanism that bacteria utilize to regulate different cellular functions, such as stress response and pathogenesis. STKs are more promiscuous than the TCS-associated kinases and can phosphorylate different protein substrates producing pleiotropic effects [16,51,52]. STKs are also able to interact with TCSs by direct phosphorylation of RRs, as reviewed in [3,53]. STK-mediated RR activation takes place on either serine or threonine residues, instead of aspartate, which is the typical residue target for HK phosphorylation. STK-mediated phosphorylation on DNA-binding domains of RR have been reported, as described for GraR in S. aureus [54], YvcK in Listeria monocytogenes [55] and RitR in S. pneumoniae [56]. STKs may also phosphorylate on receiver domains, as observed for CovR in S. pyogenes [57], WalR in B. subtillis [58], DosR in M. tuberculosis [59], or in both domains, as demonstrated for VraR in S. aureus [60].

ComE is the most studied RR in S. pneumoniae and belongs to the AlgR/AgrA/LytTR transcription factor subfamily, showing a typical receiver domain and a DNA-binding (or LytTR) domain. When phosphorylated by the ComD histidine kinase at the Asp58 residue, ComE undergoes conformational changes that increase their DNA affinity and modify transcription regulation of competence genes by binding to their promoter regions [2,47,61]. In the present work, we show that S. pneumoniae utilizes an alternative signal transduction pathway to control acidic stress response (ASIL and ATR), oxidative stress, cell wall biosynthesis, and intracellular survival in pneumocytes. Under acidic conditions, a phosphorylation crosstalk between StkP and ComE involving phosphorylation at Thr128 in the receiver domain resulted in activation of this RR.

Using the crystal structure of ComE [43], a molecular dynamic simulation of ComE permitted a comparison with the phosphomimetic ComET128E protein, predicting that the Thr128 phosphorylation produces structural changes in the DNA-binding domain. These putative conformational changes were confirmed by limited proteolysis assays that revealed differences between ComE and ComET128E. In this sense, we observed that phosphorylation at either Asp58 or Thr128 increases the dimerization and DNA-binding capacity of ComE. These results were coincident with the activation model proposed by Boudes et al [43], where the most plausible activation mechanism of ComE is first a phosphorylation reaction to induce its dimerization, which occurs at the canonical receiver domain of ComE, followed by binding to DNA via the LytTR domain. It remains to be elucidated how phosphorylation at alternative sites in the receiver domain (Asp58 or Thr128) modifies the DNA-binding domain of ComE.

Because ComE Thr128 activation is independent of CSP/ComD activation by a quorum sensing mechanism, this RR requires alternative factors to act as a sensor and/or an environmental signal to trigger an adaptive stress response. We propose that StkP senses an alternative environmental signal, acidic pH. Our results are consistent with such notion: the level of comE transcripts induced under acidic conditions is the indicator of ComE activation due to the comCDE operon is autoregulated.

An additional aspect to consider in the StkP/ComE crosstalk phosphorylation is the effect of pH on protein-protein interactions. We observed that the non-phosphorylated forms of both proteins show strong interaction at acidic pH. Once StkP auto-phosphorylates, it becomes metastable complex until it dissociates from the phosphorylated ComE form. Such cycle is favored at pH 6.0 and evidence for the outcome of such cycle is shown by the fact that phosphorylation of ComE at Thr128 by StkP prevents further interaction between these two proteins. We propose the following cycle for StkP/ComE interaction:

Pneumococcal H2O2 production is one of the most significant among bacterial pathogens, and S. pneumoniae utilizes this intermediate metabolite to compete with the respiratory tract microbiota and to produce cytotoxic effects on the host tissues [6264]. In this investigation, the transcriptome analysis of the comET128A mutant revealed a marked decrease in the expression of spxB and sodA when cells were grown under acidic conditions. These results correlated with very low H2O2 production by the comET128A mutant associated with low expression of SpxB and SodA. Considering that H2O2 is toxic for eukaryotic cells, we propose that reduction H2O2 production in the comET128A mutant facilitates its intracellular survival in pneumocytes. S. pneumoniae generates hyper-virulent mutants with defective spxB during infection [65], supporting the hypothesis that the H2O2 levels are controlled during pneumococcal pathogenesis.

Because S. pneumoniae lacks catalase, and H2O2 overproduction must be controlled for this pathogen to survive, this pathogen induces an oxidative stress resistance that is induced by endogenous H2O2 [66]. In this sense, the transcriptome analyses of the comET128A mutant revealed a decreased expression of tpxD, which encodes the thiol peroxidase. These findings correlate with the increased H2O2 susceptibility by the comET128A mutant. Similarly, a previous study showed that a tpxD (or psaD) mutant eliminated the H2O2-mediated response to high H2O2 levels [67]. Previously, a microarray analysis of the stkP mutant revealed that tpxD expression is repressed, which is correlated with a low H2O2 resistance of this mutant, but the putative regulatory mechanism was not mentioned [7]. In the present study, we have shown for the first time that the StkP/ComE pathway controls oxidative stress resistance and H2O2 production under acidic conditions, which are probably responsible for the intracellular survival of S. pneumoniae in pneumocytes.

In a previous study, we proposed that ASIL may be activated under acidic conditions by a translocation of LytA from an intracellular to an extracellular compartment probably due to cell wall alterations by an unknown ComE-dependent mechanism [68]. Here, the comET128A mutant displayed a decreased expression of murN, which encodes one of the first enzymes involved in the cell wall biosynthesis of S. pneumoniae [69]. Filipe et al [70] described that the murMN mutant showed an increased susceptibility to lysis when murMN cells were exposed to cell wall antibiotics, such as fosfomycin and vancomycin, which are involved in the inhibition of both the early and late stages of cell wall biosynthesis, respectively. Following this line of thinking, an altered expression of murN in the comET128A mutant should cause a misbalance in the peptidoglycan biosynthesis and modify susceptibility to cell wall antibiotics. Accordingly, we observed that this mutant had an increased MIC of fosfomycin compared with wt, as well as a greater tolerance to autolysis induced by fosfomycin or vancomycin. The putative cell wall alterations indicated by antibiotic susceptibility tests may explain the autolysis inhibition shown by the comET128A mutant under acidic stress, which probably interfered with LytA activation. Work is in progress to try to determine the nature of such cell wall alterations.

We also demonstrated that Thr128 phosphorylation is not involved in competence. Regarding this topic, Guiral et al [71] described a phenomenon of lysis of non-competent cells triggered by competent cells, named allolysis, which involves bacteriocins and the autolysins LytA, LytC, and CbpD. Allolysis is considered to be a competence-induced mechanism of predation of non-competent cells that contributes to virulence by releasing pneumolysin [72]. Here, we show that the StkP/ComE signaling pathway can also trigger autolysis of noncompetent cells in acidic biological niches, such as inflammatory foci or endosomal compartments. This phenomenon occurred without the activation of a quorum sensing mechanism, a situation that allows bacterial cells to lyse even under low population density conditions.

StkP and ComE have already been shown to be involved in pneumococcal pathogenesis in different studies using animal models, with StkP appearing to be involved in bacterial survival in vivo [4,73]. On the other hand, ComE-mediated competence for DNA transformation has been also associated with virulence [74,75]. As mentioned above, pneumolysin release by competence-mediated autolysis was considered to be essential for pneumococcal pathogenesis [71]. Concerning the impact of StkP/ComE pathway regulation on pneumococcal pathogenesis, we propose that two different scenarios should be considered. In extracellular niches, a subpopulation of pneumococci exposed to acidic stress may cause tissue damage by overproduction of H2O2 and induction of ASIL to release pneumolysin, with this suicidal situation being promoted by StkP-mediated phosphorylation of ComE. On the other hand, the Thr128-nonphosphorylated form of ComE might facilitate pneumococcal survival at either the extracellular or the intracellular level in host tissues. We focused our attention on the first barrier that this pathogen must cross to establish an infection, and we hypothesized that intracellular survival in pneumocytes should be important for S. pneumoniae. The ΔstkP, ΔcomE, and comET128A mutants were tested in the pneumococcal infection model in A549 pneumocytes, and they revealed an increased survival compared with wt. Thus, we conclude that this survival could have been caused by increasing their capacity of ATR, decreasing the H2O2 production and modifying the cell wall biosynthesis to repress ASIL (Fig 9). Establishing how the balance between H2O2 resistance mechanism and H2O2 production affects intracellular survival is beyond the scope of this report, but this work is in progress. Finally, we propose that the StkP/ComE pathway is relevant in the genetic regulation of physiological adaptation to environmental stress, which is necessary for pneumococcal survival in pneumocytes. This is one of the first steps in the pathogenic process that S. pneumoniae must overcome to produce infection.

thumbnail
Fig 9. Proposed model for crosstalk between StkP and ComE that impacts on the acidic stress response and intracellular survival mechanisms in S. pneumoniae exposed to acidic conditions.

https://doi.org/10.1371/journal.ppat.1007118.g009

Materials and methods

Bacterial strains, plasmids, cell lines, and growth conditions

All strains, plasmids, and oligonucleotides used in this study, as well as cloning and mutagenesis procedures, are listed in the supplementary material (S1 Table). The growth conditions and stock preparation for the pneumococcal and Escherichia coli strains have been reported elsewhere [34], and the transformation assays have also been previously described [76,77].

ASIL and ATR assays

ASIL was performed as described previously [33]. Firstly, bacterial cells were grown in Todd-Hewitt/yeast extract medium. When cultures reached OD600nm ~0.3, cells were centrifuged at 10,000 g for 5 min, the pellet was resuspended in ABM pH 6.0 and cultures were re-incubated at 37°C. Autolysis was measured as a change in OD600nm at different time points over 6 h.

ATR was performed as described previously [24]. For non-acid-induced conditions, bacterial cells were first grown in THYE (pH 7.8) at 37°C, and when cultures reached OD600nm~ 0.3, 100 μl aliquots were taken and added to 900 μl of THYE (pH 4.4) and incubated for 2 h at 37°C. Then, serial dilutions were made in THYE (pH 7.8) and plated onto 5% of sheep blood tryptic-soy agar (TSA) plates. After 24 h of incubation at 37°C, colonies were counted to determine the number of survivors, with the total CFU being obtained by plating serial dilutions of cells grown THYE pH 7.8 onto 5% sheep blood TSA, made just before cells were switched to pH 4.4. In parallel, to determine survival under acidic-induced conditions, bacterial cells were grown in THYE (pH 7.8) until OD600nm ~ 0.3, centrifuged at 10,000 g for 5 min, resuspended in THYE (pH 6.0) and incubated for 2 h at 37°C. Culture aliquots were taken and serially diluted in THYE pH 7.8 for total cell counting, while other aliquots were diluted ten times in THYE (pH 4.4) and incubated for 2 h at 37°C to determine the survival percentage as described above. For both assays (acid-induced and non acid-induced conditions), this was calculated by dividing the number of survivors at pH 4.4 by the number of total cells at time zero (before incubation at pH 4.4). For the ASIL and ATR assays, data were expressed as the mean percentage ± standard deviation (SD) of independent experiments performed in triplicate.

Cell lines and culture conditions

The A549 cell line (human lung epithelial carcinoma, pneumocytes type II; ATCC CCL-185) was cultured at 37°C, 5% CO2 in Dulbecco’s modified Eagle medium (DMEM) with 4.5 g/l of glucose and 10% of heat-inactivated fetal bovine serum (FBS)(Gibco BRL, Gaithersburg, Md.). Fully confluent A549 cells were split once every two or three days via trypsin/EDTA treatment and diluted in fresh media before being cultivated in Filter cap cell flasks of 75 cm2 (Greiner Bio-one no. 658175) until passage 6.

In vitro phosphorylation assays

In vitro phosphorylation was carried out with 0.5 μg of purified recombinant substrate protein and 0.5 μg of purified GST-StkP in 30 μl of kinase buffer (50 mM Tris-HCl, 5 mM MgCl2, 100 μM ATP, 1mM DTT, pH 7.5). The reaction was started by the addition of ATP and stopped after 60 min of incubation at 37°C by the addition of 5x Laemmli SDS sample buffer. Samples were separated by standard Tris-glycine-SDS polyacrylamide gel electrophoresis (PAGE) gels and electroblotted onto a nitrocellulose membrane. Phosphorylated proteins were detected with an anti-phosphothreonine polyclonal antibody (1∶1,000; Cell Signaling) and a goat anti-rabbit immunoglobulin G secondary antibody conjugated to horseradish peroxidase (1∶2,500; Invitrogen). Detection was performed with an enhanced chemiluminescence substrate (SuperSignal West Pico Chemiluminescent Substrate; Pierce) and Hyperfilm CL film (GE) using exposures of between 1 and 10 min. The pRSET-divIVAspn plasmid was generously provided by Dr. Orietta Massidda (Università degli studi di Cagliari, Italy) [78].

In vivo phosphorylation assays

The RC838 (comE-his) and RC839 (ΔstkP comE-his) strains (S1 Table) were grown in 2 l of ABM (pH 7.8) at 37°C until OD600nm 0.3. Half of the cultures (1 l) were centrifuged for 10 min at 5,000 x g, snap-frozen in liquid-air and stored at -80°C. The remaining 1 l was centrifuged as before, resuspended in ABM (pH 6.0), incubated at 37°C for 10 min, and finally centrifuged for 10 min at 5,000 x g, snap-frozen in liquid-air and stored at -80°C. Cell pellets were thawed in ice and resuspended in 10 ml of L8 buffer (100 mM NaH2PO4/10 mM Tris·HCl, pH 8.0/150 mM NaCl/20 mM imidazole/20mM PMSF/8 M urea) supplemented with MS-SAFE protease/phosphatase inhibitor cocktail (Sigma-Aldrich) and lysed by stirring for 1 h followed by sonication. The lysate was cleared by centrifugation at 15,000 g for 20 min, and the supernatant was added to a column packed with to 0.5 ml of Ni-NTA resin (Qiagen) equilibrated in L8 buffer. The column was washed sequentially with 10 ml of LX buffer (L buffer with X = 8, 6, 4, 2, and 0 M urea), and bound ComE-His6x protein was eluted with 2 ml of L0 buffer containing 500 mM imidazole. Protein samples (approximately 0.5 μg ComE-His) were separated by SDS-PAGE, and the gels were stained with ProQ Diamond (Invitrogen) to detect phosphorylated ComE-His, followed by SYPRO Ruby (Invitrogen) total protein staining. Gels were imaged under fluorescence mode in a Typhoon FLA 9500 scanner (GE) and protein bands were quantified using ImageQuant software (GE).

Identification of phosphorylated residues by nano-LC-MS/MS analysis

Identification of the phosphorylation site was carried out by nano-LC-MS/MS analysis as previously described [79]. Protein bands were in-gel-digested overnight with sequencing grade trypsin (Promega) at 37°C, desalted using micro-reverse phase columns (C18 Omix tips, Varian), vacuum dried and resuspended in 0.1% formic acid (v/v) in water. Tryptic peptides were injected into a nano-HPLC system (Proxeon Easy nLC, Thermo) fitted with a trap column (Easy-column C18 2 cm x 100 um ID). Posteriorly, the samples were separated on a reverse phase nano-column (Easy-Column C18 10 cm x 75 um ID; Thermo) using a linear gradient of acetonitrile 0.1% formic acid (0–45% in 70 min) at a flow rate of 400 nL/min. Mass analysis was performed using a linear ion trap mass spectrometer (LTQ Velos, Thermo) in a data-dependent mode (full scan followed by MS/MS of the top 5 peaks)[79]. Raw data was analyzed using the Proteome Discoverer software package (v.1.3.0.339, Thermo), and Sequest search engine, with the following parameters: enzyme: trypsin; maximum missed cleavage: 2; precursor mass tolerance: 1 Da; fragment mass tolerance: 0.8 Da; Ser/Thr/Tyr phosphorylation and methionine oxidation as dynamic modifications. Searches were performed using a Streptococcus pneumoniae database downloaded from UniProt (17/5/2017) and including the His tag-ComE sequence. For phosphosite localization, the phosphoRS algorithm was used and the spectra of phosphorylated peptides were manually inspected to corroborate the phosphosite assignment [80].

EMSA

The promoter region of comCDE (255 bp; pcomC) was PCR-amplified using the 5’-Cy5 labeled oligonucleotides NGEP516 and NGEP517 (IDT) and purified using the QIAquick PCR Purification Kit (QIAGEN). DNA-binding assays were performed in a total volume of 10 μl containing 50 mM NaCl, 50 mM Tris/HCl pH 7.5, 5% (v/v) glycerol, 7.5 nM Cy5-labeled PCR fragments, 1 mM MgCl2, 0.15 mg Poly(dI-dC) (as the non-specific competitor), and varying concentrations of untreated or StkP-treated ComE proteins. In the latter case, phosphorylation of ComE-Hisx6 by StkP was carried out with an equimolar amount of StkP in kinase buffer (50 mM Bis-Tris propane, 5 mM MgCl2, 1mM DTT, 0.1 μM ATP, pH 7.8 or pH 6.0) for 60 min at 37°C. Protein-DNA binding reactions were incubated at room temperature for 30 min, and “frozen” with an equal volume of 2X Stop Solution [40% (v/v) Triethylene Glycol, 10 mM Tris, pH 7.5]. DNA-protein complexes were resolved by electrophoresis in native Tris-Borate-EDTA polyacrylamide gels [10% (w/v)] containing 30% Triethylene Glycol. Gels were run at 4°C for 120 min at constant voltage (25 V cm-1) in a 0.5X TBE buffer and scanned in a Typhoon FLA 9500 biomolecular imager (GE) under fluorescence mode. Free and protein-bound DNA were quantified using ImageQuant (GE). The fraction of DNA bound (FB) at each ComE concentration was fit with a standard binding isotherm using Kaleidagraph (Synergy Software), according to the equation: FB = [ComE]/(Kd +[ComE]), where Kd is the apparent equilibrium dissociation constant and reflects the protein concentration required to shift 50% of the labeled DNA fragment.

Molecular dynamics simulation

MD simulations were carried out with the NAMD program [81], using the CHARMM27 force field [82]. Spherical boundary conditions and a non-bonded cut-off of 12.0 Å with a switching function of 10.0 Å were used. All systems were submitted to structural minimization in vacuum, and then embed in a water sphere for the MD. The temperature was set to 310 K by a Langevin thermostat. MD simulations were run for 40 ns with an integration step of 2 fs. Analysis of the trajectories was performed using VMD software [83]. Crystal structure images were analyzed using PyMOL [84].

StkP and ComE interaction by FLISA

StkP and ComE binding interactions were assessed by a sandwich fluorescence-linked immunosorbent assay (FLISA) in black 96-well high binding capacity microplates with clear flat-bottoms (Corning #3601). Each well was filled with 50 μL of 10 μg/ml GST-StkP (500 ng of GST-StkP) dissolved in a 0.1 M coating buffer (0.1 M NaHCO3/Na2CO3, pH 9.4) and incubated overnight at 4°C to allow protein adsorption. Wells were rinsed five times with Tris-buffered saline, 0.05% Tween 20, pH 7.4 (TBS-T) and the reactive sites were blocked with 2% w/v bovine serum albumin dissolved in TBS-T for 2 h at room temperature. Wells were washed three times with TBS-T. Different amounts of ComE-Hisx6 (200–1200 ng) were dissolved in 50 μL of 50 mM Bis-Tris-Propane-HCl, 1 mM MgCl2, pH 7.8, or in the same buffer but at pH 6.0, and added to StkP-coated wells and incubated for 1h at 37°C. Wells were washed 5 times with TBS-T and incubated for 1h at RT with 50 μL of a Dylight 650-conjugated anti-6X His antibody (Invitrogen MA1-21315-D650) diluted 100-fold in TBS-T. After 5 washes with TBS-T, plates were read in a Typhoon FLA 9500 scanner (GE) under fluorescence mode. Fluorescence (F) was fit using a Kaleidagraph to a standard binding isotherm with the form F = Fmax [ng ComE/(K1/2 + ng ComE)], where Fmax is the maximum fluorescence at binding saturation and reflects the maximum binding capacity (Bmax), and K1/2 is the amount of ComE (ng) required to reach half Fmax. The inverse of K1/2 represents an estimate of ComE affinity for the StkP binding sites.

Protein expression and purification

The comE gene was amplified from R801 genomic DNA with the primer pair FhkE/RhkE and cloned into the BamHI/EcoRI sites of the pRSET-A expression plasmid (Invitrogen), yielding pRSET-ComE. stkP and stkP-KD (kinase domain, amino acids 1–282) were amplified with primer pairs FstkP-ex/Rstk-ex and FstkP-ex/ Rstk-kd, respectively, and cloned into BamHI/EcoRI sites of pGEX-4T1 expression plasmid (GE) to generate pGEX-StkP and pGEX-StkP-KD. Plasmid pTrc-LytA(N) expressing the amino-terminal region of LytA (1–206) was obtained by cloning a lytA fragment generated by PCR amplification with primers FlytA1/RlytA2 into the BamHI/EcoRI sites of pTrcHis2A (Invitrogen). Mutations were introduced in pRSET-ComE by Quickchange site-directed mutagenesis (Agilent), employing primer pairs NGEP514/NGEP515, NGEP75/NGEP76 and NGEP77/NGEP78 to obtain pRSET-ComE(D58E), pRSET-ComE(T128A), and pRSET-ComE(T128E), respectively. In the same way, the K42M mutation was introduced in pGEX-StkP with primers NGEP770/771 to give pGEX-StkP(K42M).

Soluble His6X-tagged LytA(N) and ComE-Hisx6 proteins were purified from the E. coli BL21(DE3) strain co-transformed with either pTrc-LytA(N) or pRSet-ComE derivatives and chaperone expression plasmids pBB540 and pBB550 [2,85]. E. coli cells were grown on 800 ml of Terrific broth and induced with 100 μM IPTG according to de Marco [85]. His-tagged proteins were purified from protein lysates obtained by sonication using an NTA-Ni2+ resin (Qiagen) following the manufacturer’s protocol. Eluted protein was further purified by gel filtration using a HiPrep 16/60 Sephacryl S-200 HR column mounted in a ÄKTA purifier system (GE). ComE containing fractions were pooled, concentrated with an Amicon Ultra-4 centrifugal filter (Millipore), and dialyzed against the storage buffer [50 mM Tris, 200 mM NaCl, 1mM DTT, 50% v/v glycerol, pH 7.5). Samples were snap frozen and stored at -80°C until use. Following exactly the same protocol as above, GST-tagged StkP proteins were purified from the soluble protein fraction of BL21(DE3) cells bearing pGEX-StkP derivatives and plasmids pBB540 and pBB50. In this case, a Glutathione Sepharose resin (GE) was used to retain GST-tagged StkP. DivIVA was purified from BL21 (DE3) cells transformed with pRSET-divIVA according to Fadda et al. [78]. Purified recombinant Aequorea victoria His-tagged GFP protein was purchased from SIGMA.

Dimeric state of ComE proteins

Native PAGE was used to assess the ComE monomer/dimer ratio. Purified ComE-Hisx6 proteins were diluted in 2x Laemmli sample buffer without 2-mercaptoethanol and SDS and loaded in a 4–20% gradient Bis-Tris precast polyacrylamide gel (GenScript). Electrophoresis was performed at 4°C using Tris-MOPS running buffer without SDS (GenScript) at a constant electric field of 15 V cm-1. Proteins were electroblotted onto a PVDF membrane and probed with Dylight 650-conjugated anti-6X His antibody to detect His-tagged ComE. The membranes were imaged under fluorescence mode in a Typhoon FLA 9500 scanner (GE Healthcare), and bands were quantified with ImageQuant software (GE Healthcare).

RNAseq analysis

Cells were initially grown in THYE medium at pH 7.8 until OD600nm ~0.3 (log phase), centrifuged at 14,000 g for 10 min at 4°C, resuspended in the same volume in ABM at pH 6.0 (Piñas et al, 2008) and incubated a 37°C for 1h. Then, cells were centrifuged at 14,000 x g for 10 min at 4°C, resuspended in a 1/10 vol of lysis buffer (DOC 1% in 0.9% Na Cl) and incubated 3 min a 37°C until complete lysis. Total RNA was purified by TRIzol reagent according to the manufacturer's instructions (Fisher Scientific) from three biological replicates for wt and the comET128A mutant. Posteriorly, we used the Ribopure Bacterial RNA Purification Kit (Ambion) following the manufacturer's protocol, with the contaminant DNA being removed using the provided Dnase. rRNA was depleted from 8μg of total RNA using the MICROBExpress Bacterial mRNA Enrichment Kit (Ambion), and then the transcriptome libraries were prepared with TruSeq Stranded RNA Library Preparation Kit (Illumina) following the manufacturer's instructions. Briefly, enriched mRNA was fragmented using reagents provided with the kit, and this was followed by first-strand cDNA synthesis and second-strand generation. The libraries were tagged with unique indexes and amplified for a limited number of PCR cycles followed by quantification and qualification using the DNA High Sensitivity Assay Kit. Samples were sequenced using PE150bp chemistry and the Illumina HiSeq. Reads were trimmed by Trimmomatic 0.36 [86] to generate high-quality reads. Subsequently, these reads of wt and the comET128A samples were separately aligned to the Streptococcus pneumoniae R6 genome using BWA -version 0.7.12-r1039 (bio-bwa.sourceforge.net) at default parameters. The software package SAMtools (http://samtools.sourceforge.net/) was used to convert the sequence alignment/map (SAM) file to a sorted binary alignment/map (BAM) file. The mapped reads ratio (MRR) to the reference in each dataset was calculated by applying the flagstat command of SAMtools software to the BAM file.

Differential gene expression

The aligned reads were assembled by Cufflinks (version-2.2.1), and then the differentially expressed genes were detected and quantified by Cuffdiff, which is included in the Cufflinks package, using a rigorous sophisticated statistical analysis. The expression of the genes was calculated in terms of FPKM (Fragment per kilobase per million mapped reads). Differential gene expression analysis was carried out between wt and the comET128A samples.

qRT-PCR

cDNA was synthesized from 2 μg RNA using the ProtoScript II First Strand cDNA Synthesis Kit (NEB) following the manufacture's protocol. cDNA was cleaned using the QIAquick PCR Purification Kit (Qiagen). Genes were amplified using the oligos listed in the S2 Table and FastStart Essential DNA Green Master Mix (Roche) following the manufacturer's protocol. Expression was determined relative to AU0158 normalized by gyrA (spr1099) expression using the ΔΔCt method [87]. The gyrA had a similar expression by RNA-Seq for wt and the comET128A mutant, and this had been used to normalize the expression in S. pneumoniae in other studies [88].

Intracellular survival assays

The assays to determine the intracellular survival of pneumococci were performed as reported previously [33], but with modifications. Briefly, 3.0 × 105 of A549 cells per well were seeded in 6 well plates and cultured in DMEM supplemented with 10% of fetal bovine serum (FBS) and incubated for 12 h. Pneumococci were grown in THYE to the mid-log phase (OD600nm 0.3) and resuspended in DMEM (with 10% FBS). Infection of cell monolayers was carried out using a multiplicity of infection (MOI) 20:1. Bacterial internalization after incubation and washes with extracellular antibiotics was approximately 1%, and the occurrence of apoptosis/necrosis caused by pneumococcal infection quantified by flow cytometry (Annexin V/propidium iodide labeling kit; Invitrogen) was approximately 5–10% for all time points analyzed. A549 cells were incubated 3 h with pneumococcal strains and cells were washed three times with phosphate-buffered saline (PBS) and fresh DMEM (without FBS) containing 150 μg/ml potassium penicillin G (Sigma P7794) and 900 μg/ml gentamicin sulfate (US Biological G2030). After a 20 min rest period, cells were washed three times with PBS. The eukaryotic cells were lysed by centrifugation for 5 min at 10,000 rpm and the bacterial pellet was resuspended in THYE medium. The number of internalized bacteria at different time points was quantified after serial dilutions and plating on BHI 5% sheep blood agar plates with incubation for 16 h at 37°C. The time scale referred to the time after elimination of the extracellular bacteria by antibiotic treatment. A 100% survival was defined after 20 min of antibiotic treatment (S1 Fig), and all the samples were referred to this point to calculate the respective percentages.

Hydrogen peroxide determination

For the detection of H2O2 released by bacterial cells, the phenol red oxidation microassay was used. Briefly, cells were grown in BHI to the mid-log phase (OD600nm 0.3). Posteriorly, cells were centrifuged at 10,000 x g for 5 min, resuspended in Todd Hewitt broth THB (pH 6.0) and incubated by 1 h at 37°C. Aliquots were taken and serially diluted to determine viable cells by plating in BHI-blood agar. Other aliquots were centrifuged at 10,000 x g for 5 min, and 100 μl of supernatants were transferred to multiwell plates and mixed with the same volume of PRS buffer (NaCl 140 mM, dextrose 5.5 mM, phenol red 280 μM, and horseradish peroxidase 8.5 U/ml in phosphate-buffered saline, pH7.0). Reactions were incubated for 90 min at 37°C and the reaction was stopped with 10 μl of 1 N NaOH, and the reactive wells were read in a microplate reader (Bio-Rad) with a 595-nm filter. Assays were performed in triplicate and results are expressed as mmoles of H2O2 released by 106 cells.

Hydrogen peroxide susceptibility assays

Bacterial strains were grown until OD600nm in BHI and aliquots were treated with H2O2 20 mM (final concentration). Every 30 min, aliquots were taken and serially diluted to determine viable cells by plating in BHI-blood agar. The percent survival was calculated by dividing the CFU of cultures after exposure to H2O2 by the CFU of the control tube without H2O2. Assays were performed in triplicate and results are shown survival percentage at different time points.

Limited proteolysis assays

Limited proteolysis with proteinase K was carried out in a 30 μl reaction volume with 3 μg of ComE or ComET128E and 6 ng of the proteinase K in 10 mM Tris, 1 mM CaCl2, pH 7.5, for 30 min at room temperature. Reactions were stopped with 5 mM PMSF, 5 mM EDTA and 1X Laemmli loading buffer, and immediately boiled for 5 min. Digestions with trypsin were performed under the same conditions as before but in 100 mM Tris, pH 8.5. Trypsinized samples were boiled immediately for 5 min after stopping the reactions with 5 mM PMSF and 1X Laemmli loading buffer. The extension of protein digestion was verified by 12% SDS-PAGE followed by SYPRO Ruby staining.

Accession numbers

The RNA-seq data generated from this study are deposited at the NCBI SRA under the accession numbers SAMN08473835 and SAMN08473836.

Supporting information

S1 Fig. Determination of CSP-induced transformability in S. pneumoniae mutants.

https://doi.org/10.1371/journal.ppat.1007118.s001

(TIF)

S2 Fig. Phenotypical characterization of the stkPK42R and ΔmapZ mutants.

https://doi.org/10.1371/journal.ppat.1007118.s002

(TIF)

S3 Fig. StkP and ComE mediate pcomC activation by acidic stress.

https://doi.org/10.1371/journal.ppat.1007118.s003

(TIF)

S4 Fig. (video). Thr128 phosphorylation changes the DNA-binding domain of ComE.

https://doi.org/10.1371/journal.ppat.1007118.s004

(MPG)

S5 Fig. Conformational changes between ComE and ComET128E determined by limited proteolysis assays.

https://doi.org/10.1371/journal.ppat.1007118.s005

(TIF)

S6 Fig. StkP-mediated phosphorylation of ComE is dependent on pH and alters its DNA-binding affinity.

https://doi.org/10.1371/journal.ppat.1007118.s006

(TIF)

S7 Fig. Kinase activity of StkP modifies the ComE phosphorylation and alters EMSA.

https://doi.org/10.1371/journal.ppat.1007118.s007

(TIF)

S8 Fig. In vitro interactions between StkP and ComE.

https://doi.org/10.1371/journal.ppat.1007118.s008

(TIF)

S9 Fig. Putative DNA binding motif in ComE-regulated genes.

https://doi.org/10.1371/journal.ppat.1007118.s009

(TIF)

S1 Table. Plasmids and strains used in this work.

https://doi.org/10.1371/journal.ppat.1007118.s010

(DOCX)

S2 Table. Evaluation of acidic-stress induced lysis in hk mutants.

https://doi.org/10.1371/journal.ppat.1007118.s011

(DOCX)

S3 Table. List of ComE-regulated genes as determined by RNAseq analysis.

https://doi.org/10.1371/journal.ppat.1007118.s012

(XLSX)

Acknowledgments

We thank Don Morrison (the University of Illinois at Chicago, USA) for providing us with the Janus cassette, Orietta Massidda (Università degli Studi di Cagliari, Italy) for providing the pRSET-divIVAspn plasmid, Gustavo Bonacci and Juan Nicola for technical discussions. We also thank native speaker Dr. Paul Hobson (Cultura Britanica-Argentina) for revising this manuscript.

References

  1. 1. Salazar ME, Laub MT (2015) Temporal and evolutionary dynamics of two-component signaling pathways. Curr Opin Microbiol 24: 7–14. pmid:25589045
  2. 2. Claverys JP, Prudhomme M, Martin B (2006) Induction of competence regulons as a general response to stress in gram-positive bacteria. Annu Rev Microbiol 60: 451–475. pmid:16771651
  3. 3. Dworkin J (2015) Ser/Thr phosphorylation as a regulatory mechanism in bacteria. Curr Opin Microbiol 24: 47–52. pmid:25625314
  4. 4. Echenique J, Kadioglu A, Romao S, Andrew PW, Trombe MC (2004) Protein serine/threonine kinase StkP positively controls virulence and competence in Streptococcus pneumoniae. Infect Immun 72: 2434–2437. pmid:15039376
  5. 5. Pallova P, Hercik K, Saskova L, Novakova L, Branny P (2007) A eukaryotic-type serine/threonine protein kinase StkP of Streptococcus pneumoniae acts as a dimer in vivo. Biochemical and Biophysical Research Communications 355: 526–530. pmid:17307148
  6. 6. Yeats C, Finn RD, Bateman A (2002) The PASTA domain: a beta-lactam-binding domain. Trends in Biochemical Sciences 27: 438. pmid:12217513
  7. 7. Saskova L, Novakova L, Basler M, Branny P (2007) Eukaryotic-type serine/threonine protein kinase StkP is a global regulator of gene expression in Streptococcus pneumoniae. Journal of Bacteriology 189: 4168–4179. pmid:17416671
  8. 8. Fleurie A, Cluzel C, Guiral S, Freton C, Galisson F, et al. (2012) Mutational dissection of the S/T-kinase StkP reveals crucial roles in cell division of Streptococcus pneumoniae. Mol Microbiol 83: 746–758. pmid:22211696
  9. 9. Fleurie A, Lesterlin C, Manuse S, Zhao C, Cluzel C, et al. (2014) MapZ marks the division sites and positions FtsZ rings in Streptococcus pneumoniae. Nature 516: 259–262. pmid:25470041
  10. 10. Stamsas GA, Straume D, Ruud Winther A, Kjos M, Frantzen CA, et al. (2017) Identification of EloR (Spr1851) as a regulator of cell elongation in Streptococcus pneumoniae. Mol Microbiol 105: 954–967. pmid:28710862
  11. 11. Novakova L, Bezouskova S, Pompach P, Spidlova P, Saskova L, et al. (2010) Identification of multiple substrates of the StkP Ser/Thr protein kinase in Streptococcus pneumoniae. Journal of Bacteriology 192: 3629–3638. pmid:20453092
  12. 12. Giefing C, Jelencsics KE, Gelbmann D, Senn BM, Nagy E (2010) The pneumococcal eukaryotic-type serine/threonine protein kinase StkP co-localizes with the cell division apparatus and interacts with FtsZ in vitro. Microbiology 156: 1697–1707. pmid:20223804
  13. 13. Beilharz K, Novakova L, Fadda D, Branny P, Massidda O, et al. (2012) Control of cell division in Streptococcus pneumoniae by the conserved Ser/Thr protein kinase StkP. Proc Natl Acad Sci U S A 109: E905–913. pmid:22431591
  14. 14. Manuse S, Fleurie A, Zucchini L, Lesterlin C, Grangeasse C (2016) Role of eukaryotic-like serine/threonine kinases in bacterial cell division and morphogenesis. FEMS Microbiol Rev 40: 41–56. pmid:26429880
  15. 15. Giefing C, Meinke AL, Hanner M, Henics T, Bui MD, et al. (2008) Discovery of a novel class of highly conserved vaccine antigens using genomic scale antigenic fingerprinting of pneumococcus with human antibodies. Journal of Experimental Medicine 205: 117–131. pmid:18166586
  16. 16. Grangeasse C (2016) Rewiring the Pneumococcal Cell Cycle with Serine/Threonine- and Tyrosine-kinases. Trends Microbiol.
  17. 17. Herbert JA, Mitchell AM, Mitchell TJ (2015) A Serine-Threonine Kinase (StkP) Regulates Expression of the Pneumococcal Pilus and Modulates Bacterial Adherence to Human Epithelial and Endothelial Cells In Vitro. PLoS One 10: e0127212. pmid:26090876
  18. 18. Martin B, Granadel C, Campo N, Henard V, Prudhomme M, et al. (2010) Expression and maintenance of ComD-ComE, the two-component signal-transduction system that controls competence of Streptococcus pneumoniae. Mol Microbiol 75: 1513–1528. pmid:20180906
  19. 19. Claverys JP, Havarstein LS (2002) Extracellular-peptide control of competence for genetic transformation in Streptococcus pneumoniae. Frontiers in Bioscience 7: d1798–1814. pmid:12133809
  20. 20. Havarstein LS, Coomaraswamy G, Morrison DA (1995) An unmodified heptadecapeptide pheromone induces competence for genetic transformation in Streptococcus pneumoniae. Proceedings of the National Academy of Sciences of the United States of America 92: 11140–11144. pmid:7479953
  21. 21. Lee MS, Morrison DA (1999) Identification of a new regulator in Streptococcus pneumoniae linking quorum sensing to competence for genetic transformation. Journal of Bacteriology 181: 5004–5016. pmid:10438773
  22. 22. Osaki M, Arcondeguy T, Bastide A, Touriol C, Prats H, et al. (2009) The StkP/PhpP signaling couple in Streptococcus pneumoniae: cellular organization and physiological characterization. Journal of Bacteriology 191: 4943–4950. pmid:19502404
  23. 23. Lardner A (2001) The effects of extracellular pH on immune function. J Leukoc Biol 69: 522–530. pmid:11310837
  24. 24. Grinstein S, Swallow CJ, Rotstein OD (1991) Regulation of cytoplasmic pH in phagocytic cell function and dysfunction. Clin Biochem 24: 241–247. pmid:1651820
  25. 25. Borregaard N, Herlin T (1982) Energy metabolism of human neutrophils during phagocytosis. J Clin Invest 70: 550–557. pmid:7107894
  26. 26. Roiniotis J, Dinh H, Masendycz P, Turner A, Elsegood CL, et al. (2009) Hypoxia prolongs monocyte/macrophage survival and enhanced glycolysis is associated with their maturation under aerobic conditions. J Immunol 182: 7974–7981. pmid:19494322
  27. 27. Light RW, Girard WM, Jenkinson SG, George RB (1980) Parapneumonic effusions. Am J Med 69: 507–512. pmid:7424940
  28. 28. Bassoe CF, Bjerknes R (1985) Phagocytosis by human leukocytes, phagosomal pH and degradation of seven species of bacteria measured by flow cytometry. J Med Microbiol 19: 115–125. pmid:3968704
  29. 29. Ring A, Weiser JN, Tuomanen EI (1998) Pneumococcal trafficking across the blood-brain barrier. Molecular analysis of a novel bidirectional pathway. J Clin Invest 102: 347–360. pmid:9664076
  30. 30. Radin JN, Orihuela CJ, Murti G, Guglielmo C, Murray PJ, et al. (2005) Beta-Arrestin 1 participates in platelet-activating factor receptor-mediated endocytosis of Streptococcus pneumoniae. Infect Immun 73: 7827–7835. pmid:16299272
  31. 31. Huotari J, Helenius A (2011) Endosome maturation. EMBO J 30: 3481–3500. pmid:21878991
  32. 32. Martin-Galiano AJ, Overweg K, Ferrandiz MJ, Reuter M, Wells JM, et al. (2005) Transcriptional analysis of the acid tolerance response in Streptococcus pneumoniae. Microbiology 151: 3935–3946. pmid:16339938
  33. 33. Cortes PR, Pinas GE, Cian MB, Yandar N, Echenique J (2015) Stress-triggered signaling affecting survival or suicide of Streptococcus pneumoniae. Int J Med Microbiol 305: 157–169. pmid:25543170
  34. 34. Pinas GE, Cortes PR, Orio AG, Echenique J (2008) Acidic stress induces autolysis by a CSP-independent ComE pathway in Streptococcus pneumoniae. Microbiology 154: 1300–1308. pmid:18451038
  35. 35. Johnsborg O, Havarstein LS (2009) Regulation of natural genetic transformation and acquisition of transforming DNA in Streptococcus pneumoniae. FEMS Microbiol Rev 33: 627–642. pmid:19396959
  36. 36. Kalantari A, Derouiche A, Shi L, Mijakovic I (2015) Serine/threonine/tyrosine phosphorylation regulates DNA binding of bacterial transcriptional regulators. Microbiology 161: 1720–1729. pmid:26220449
  37. 37. Klein AH, Shulla A, Reimann SA, Keating DH, Wolfe AJ (2007) The intracellular concentration of acetyl phosphate in Escherichia coli is sufficient for direct phosphorylation of two-component response regulators. Journal of Bacteriology 189: 5574–5581. pmid:17545286
  38. 38. Goulian M (2010) Two-component signaling circuit structure and properties. Curr Opin Microbiol 13: 184–189. pmid:20149717
  39. 39. Novakova L, Saskova L, Pallova P, Janecek J, Novotna J, et al. (2005) Characterization of a eukaryotic type serine/threonine protein kinase and protein phosphatase of Streptococcus pneumoniae and identification of kinase substrates. FEBS J 272: 1243–1254. pmid:15720398
  40. 40. Martin B, Prudhomme M, Alloing G, Granadel C, Claverys JP (2000) Cross-regulation of competence pheromone production and export in the early control of transformation in Streptococcus pneumoniae. Mol Microbiol 38: 867–878. pmid:11115120
  41. 41. Pestova EV, Havarstein LS, Morrison DA (1996) Regulation of competence for genetic transformation in Streptococcus pneumoniae by an auto-induced peptide pheromone and a two-component regulatory system. Molecular Microbiology 21: 853–862. pmid:8878046
  42. 42. Lopez R, Garcia E (2004) Recent trends on the molecular biology of pneumococcal capsules, lytic enzymes, and bacteriophage. FEMS Microbiology Reviews 28: 553–580. pmid:15539074
  43. 43. Boudes M, Sanchez D, Graille M, van Tilbeurgh H, Durand D, et al. (2014) Structural insights into the dimerization of the response regulator ComE from Streptococcus pneumoniae. Nucleic Acids Res 42: 5302–5313. pmid:24500202
  44. 44. Jung K, Fried L, Behr S, Heermann R (2012) Histidine kinases and response regulators in networks. Curr Opin Microbiol 15: 118–124. pmid:22172627
  45. 45. Sidote DJ, Barbieri CM, Wu T, Stock AM (2008) Structure of the Staphylococcus aureus AgrA LytTR domain bound to DNA reveals a beta fold with an unusual mode of binding. Structure 16: 727–735. pmid:18462677
  46. 46. Pestova EV, Havarstein LS, Morrison DA (1996) Regulation of competence for genetic transformation in Streptococcus pneumoniae by an auto-induced peptide pheromone and a two-component regulatory system. Mol Microbiol 21: 853–862. pmid:8878046
  47. 47. Martin B, Soulet al, Mirouze N, Prudhomme M, Mortier-Barriere I, et al. (2013) ComE/ComE~P interplay dictates activation or extinction status of pneumococcal X-state (competence). Mol Microbiol 87: 394–411. pmid:23216914
  48. 48. Yesilkaya H, Kadioglu A, Gingles N, Alexander JE, Mitchell TJ, et al. (2000) Role of manganese-containing superoxide dismutase in oxidative stress and virulence of Streptococcus pneumoniae. Infect Immun 68: 2819–2826. pmid:10768978
  49. 49. Hajaj B, Yesilkaya H, Benisty R, David M, Andrew PW, et al. (2012) Thiol peroxidase is an important component of Streptococcus pneumoniae in oxygenated environments. Infect Immun 80: 4333–4343. pmid:23027531
  50. 50. Filipe SR, Pinho MG, Tomasz A (2000) Characterization of the murMN operon involved in the synthesis of branched peptidoglycan peptides in Streptococcus pneumoniae. J Biol Chem 275: 27768–27774. pmid:10869361
  51. 51. Shi L, Pigeonneau N, Ravikumar V, Dobrinic P, Macek B, et al. (2014) Cross-phosphorylation of bacterial serine/threonine and tyrosine protein kinases on key regulatory residues. Front Microbiol 5: 495. pmid:25278935
  52. 52. Wright DP, Ulijasz AT (2014) Regulation of transcription by eukaryotic-like serine-threonine kinases and phosphatases in Gram-positive bacterial pathogens. Virulence 5: 863–885. pmid:25603430
  53. 53. Burnside K, Rajagopal L (2012) Regulation of prokaryotic gene expression by eukaryotic-like enzymes. Curr Opin Microbiol 15: 125–131. pmid:22221896
  54. 54. Fridman M, Williams GD, Muzamal U, Hunter H, Siu KW, et al. (2013) Two unique phosphorylation-driven signaling pathways crosstalk in Staphylococcus aureus to modulate the cell-wall charge: Stk1/Stp1 meets GraSR. Biochemistry 52: 7975–7986. pmid:24102310
  55. 55. Pensinger DA, Boldon KM, Chen GY, Vincent WJ, Sherman K, et al. (2016) The Listeria monocytogenes PASTA Kinase PrkA and Its Substrate YvcK Are Required for Cell Wall Homeostasis, Metabolism, and Virulence. PLoS Pathog 12: e1006001. pmid:27806131
  56. 56. Ulijasz AT, Falk SP, Weisblum B (2009) Phosphorylation of the RitR DNA-binding domain by a Ser-Thr phosphokinase: implications for global gene regulation in the streptococci. Mol Microbiol 71: 382–390. pmid:19040630
  57. 57. Horstmann N, Saldana M, Sahasrabhojane P, Yao H, Su X, et al. (2014) Dual-site phosphorylation of the control of virulence regulator impacts group a streptococcal global gene expression and pathogenesis. PLoS Pathog 10: e1004088. pmid:24788524
  58. 58. Libby EA, Goss LA, Dworkin J (2015) The Eukaryotic-Like Ser/Thr Kinase PrkC Regulates the Essential WalRK Two-Component System in Bacillus subtilis. PLoS Genet 11: e1005275. pmid:26102633
  59. 59. Chao JD, Papavinasasundaram KG, Zheng X, Chavez-Steenbock A, Wang X, et al. (2010) Convergence of Ser/Thr and two-component signaling to coordinate expression of the dormancy regulon in Mycobacterium tuberculosis. J Biol Chem 285: 29239–29246. pmid:20630871
  60. 60. Canova MJ, Baronian G, Brelle S, Cohen-Gonsaud M, Bischoff M, et al. (2014) A novel mode of regulation of the Staphylococcus aureus Vancomycin-resistance-associated response regulator VraR mediated by Stk1 protein phosphorylation. Biochem Biophys Res Commun 447: 165–171. pmid:24704444
  61. 61. Ween O, Gaustad P, Havarstein LS (1999) Identification of DNA binding sites for ComE, a key regulator of natural competence in Streptococcus pneumoniae. Mol Microbiol 33: 817–827. pmid:10447890
  62. 62. Echlin H, Frank MW, Iverson A, Chang TC, Johnson MD, et al. (2016) Pyruvate Oxidase as a Critical Link between Metabolism and Capsule Biosynthesis in Streptococcus pneumoniae. PLoS Pathog 12: e1005951. pmid:27760231
  63. 63. Pericone CD, Overweg K, Hermans PW, Weiser JN (2000) Inhibitory and bactericidal effects of hydrogen peroxide production by Streptococcus pneumoniae on other inhabitants of the upper respiratory tract. Infect Immun 68: 3990–3997. pmid:10858213
  64. 64. Regev-Yochay G, Trzcinski K, Thompson CM, Lipsitch M, Malley R (2007) SpxB is a suicide gene of Streptococcus pneumoniae and confers a selective advantage in an in vivo competitive colonization model. J Bacteriol 189: 6532–6539. pmid:17631628
  65. 65. Syk A, Norman M, Fernebro J, Gallotta M, Farmand S, et al. (2014) Emergence of hypervirulent mutants resistant to early clearance during systemic serotype 1 pneumococcal infection in mice and humans. J Infect Dis 210: 4–13. pmid:24443543
  66. 66. Pericone CD, Park S, Imlay JA, Weiser JN (2003) Factors contributing to hydrogen peroxide resistance in Streptococcus pneumoniae include pyruvate oxidase (SpxB) and avoidance of the toxic effects of the fenton reaction. J Bacteriol 185: 6815–6825. pmid:14617646
  67. 67. Tseng HJ, McEwan AG, Paton JC, Jennings MP (2002) Virulence of Streptococcus pneumoniae: PsaA mutants are hypersensitive to oxidative stress. Infect Immun 70: 1635–1639. pmid:11854257
  68. 68. Cortes PR, Piñas GE, Cian MB, Yandar N, Echenique J (2015) Stress-triggered signaling affecting survival or suicide of Streptococcus pneumoniae. International Journal of Medical Microbiology 304.
  69. 69. Filipe SR, Tomasz A (2000) Inhibition of the expression of penicillin resistance in Streptococcus pneumoniae by inactivation of cell wall muropeptide branching genes. Proc Natl Acad Sci U S A 97: 4891–4896. pmid:10759563
  70. 70. Filipe SR, Severina E, Tomasz A (2002) The murMN operon: a functional link between antibiotic resistance and antibiotic tolerance in Streptococcus pneumoniae. Proc Natl Acad Sci U S A 99: 1550–1555. pmid:11830670
  71. 71. Guiral S, Mitchell TJ, Martin B, Claverys JP (2005) Competence-programmed predation of noncompetent cells in the human pathogen Streptococcus pneumoniae: genetic requirements. Proc Natl Acad Sci U S A 102: 8710–8715. pmid:15928084
  72. 72. Veening JW, Blokesch M (2017) Interbacterial predation as a strategy for DNA acquisition in naturally competent bacteria. Nat Rev Microbiol. 15(10):629. pmid:28736449
  73. 73. Oggioni MR, Trappetti C, Kadioglu A, Cassone M, Iannelli F, et al. (2006) Switch from planktonic to sessile life: a major event in pneumococcal pathogenesis. Mol Microbiol 61: 1196–1210. pmid:16925554
  74. 74. Zhu L, Lau GW (2011) Inhibition of competence development, horizontal gene transfer and virulence in Streptococcus pneumoniae by a modified competence stimulating peptide. PLoS Pathog 7: e1002241. pmid:21909280
  75. 75. Kowalko JE, Sebert ME (2008) The Streptococcus pneumoniae competence regulatory system influences respiratory tract colonization. Infect Immun 76: 3131–3140. pmid:18443092
  76. 76. Albarracin Orio AG, Pinas GE, Cortes PR, Cian MB, Echenique J (2011) Compensatory evolution of pbp mutations restores the fitness cost imposed by beta-lactam resistance in Streptococcus pneumoniae. PLoS Pathog 7: e1002000. pmid:21379570
  77. 77. Echenique JR, Chapuy-Regaud S, Trombe MC (2000) Competence regulation by oxygen in Streptococcus pneumoniae: involvement of ciaRH and comCDE. Mol Microbiol 36: 688–696. pmid:10844657
  78. 78. Fadda D, Santona A, D'Ulisse V, Ghelardini P, Ennas MG, et al. (2007) Streptococcus pneumoniae DivIVA: localization and interactions in a MinCD-free context. J Bacteriol 189: 1288–1298. pmid:17098892
  79. 79. Lisa MN, Gil M, Andre-Leroux G, Barilone N, Duran R, et al. (2015) Molecular Basis of the Activity and the Regulation of the Eukaryotic-like S/T Protein Kinase PknG from Mycobacterium tuberculosis. Structure 23: 1039–1048. pmid:25960409
  80. 80. Taus T, Kocher T, Pichler P, Paschke C, Schmidt A, et al. (2011) Universal and confident phosphorylation site localization using phosphoRS. J Proteome Res 10: 5354–5362. pmid:22073976
  81. 81. Phillips JC, Braun R, Wang W, Gumbart J, Tajkhorshid E, et al. (2005) Scalable molecular dynamics with NAMD. J Comput Chem 26: 1781–1802. pmid:16222654
  82. 82. MacKerell AD, Bashford D, Bellott M, Dunbrack RL, Evanseck JD, et al. (1998) All-atom empirical potential for molecular modeling and dynamics studies of proteins. J Phys Chem B 102: 3586–3616. pmid:24889800
  83. 83. Humphrey W, Dalke A, Schulten K (1996) VMD: visual molecular dynamics. J Mol Graph 14: 33–38, 27–38. pmid:8744570
  84. 84. DeLano WL (2002) The PyMOL Molecular Graphics System.
  85. 85. de Marco A (2007) Protocol for preparing proteins with improved solubility by co-expressing with molecular chaperones in Escherichia coli. Nat Protoc 2: 2632–2639. pmid:17948006
  86. 86. Bolger AM, Lohse M, Usadel B (2014) Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30: 2114–2120. pmid:24695404
  87. 87. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25: 402–408. pmid:11846609
  88. 88. Mann B, van Opijnen T, Wang J, Obert C, Wang YD, et al. (2012) Control of virulence by small RNAs in Streptococcus pneumoniae. PLoS Pathog 8: e1002788. pmid:22807675