Introduction: The hydrophilic, polymeric chain of the lipoteichoic acid (LTA) of the Gram-positive pathobiont Streptococcus pneumoniae is covalently linked to the glycosylglycerolipid α-d-glucopyranosyl-(1,3)-diacylglycerol by the LTA ligase TacL, leading to its fixation in the cytoplasmic membrane. Pneumococcal LTA, sharing identical repeating units with the wall teichoic acids (WTA), is dispensable for normal growth but required for full virulence in invasive infections. Methods: Mutants deficient in TacL and complemented strains constructed were tested for their growth, resistance against oxidative stress, and susceptibility against antimicrobial peptides. Further, the membrane fluidity of pneumococci, their capability to adhere to lung epithelial cells, and virulence in a Galleria mellonella as well as intranasal mouse infection model were assessed. Results: In the present study, we indicate that LTA is already indispensable for pneumococcal adherence to human nasopharyngeal cells and colonization in an intranasal mouse infection model. Mutants deficient for TacL did not show morphological defects. However, our analysis of pneumococcal membranes in different serotypes showed an altered membrane fluidity and surface protein abundance of lipoproteins in mutants deficient for LTA but not WTA. These mutants had a decreased membrane fluidity, exhibited higher amounts of lipoproteins, and showed an increased susceptibility to antimicrobial peptides. In complemented mutant strains, this defect was fully restored. Conclusion: Taken together, LTA is crucial for colonization and required to effectively protect pneumococci from innate immune defence mechanisms by maintaining the membrane integrity.

The human pathobiont Streptococcus pneumoniae belongs to the common microbiota of the human respiratory tract. However, S. pneumoniae (the pneumococcus) is also the aetiologic agent and leading cause of death from lower respiratory tract infections. Subpopulations of high risk are in particular the elderly as well as young children without vaccination [1]. The cell wall of S. pneumoniae is a complex construct of different polysaccharides. The outermost layer is the capsular polysaccharide (CPS), which surrounds the bacterium, thereby masking surface proteins. CPS protects the pneumococcus against external influences, avoids detection by the immune system, and represents a crucial virulence factor, e.g., by preventing phagocytosis [2, 3]. Despite the large number of more than 100 different serotypes, CPS is the primary target of most common vaccines [4‒8]. Other important macromolecules of the cell wall are teichoic acids (TAs), which offer a binding site for the family of choline-binding proteins (CBPs) through their characteristic phophorylcholine (P-Cho) decorations. CBPs are of high relevance for physiological remodelling of the cell wall (LytA, LytB, LytC), adhesion to host cells (PspC), and immune evasion (PspA) [9‒11]. In contrast to other Gram-positive bacteria, pneumococcal wall teichoic acids (WTAs) and lipoteichoic acids (LTAs) have identical repeating units and share an identical intracellular biosynthetic pathway [12‒14]. However, WTAs are anchored to the peptidoglycan (PGN), whereas LTAs are anchored in the cell membrane [12, 15]. The final ligation step to form LTA, in which the TA precursor chains are transferred to the glycosylglycerolipid α-d-glucopyranosyl-(1,3)-diacylglycerol serving as anchor of the LTA in the cell membrane, is mediated by the TA ligase TacL [14]. The cell membrane of Gram-positive bacteria consists of different molecules belonging to the families of glycerolipids and glycerophospholipids. The glycerolipids identified in S. pneumoniae are diacylglycerol, α-d-glucopyranosyl-(1,3)-diacylglycerol, and α-d-galactopyranosyl-(1,2)-α-d-glucopyranosyl-(1,3)-diacylglycerol [16, 17]. Negatively charged phosphatidylglycerol and cardiolipin are the major glycerophospholipids present in the cell membrane of S. pneumoniae [18, 19]. Due to the emergence of resistance to antibiotics, antimicrobial peptides (AMPs) targeting the cell membrane of bacteria are becoming highly attractive anti-infectives. Cationic AMPs can bind to anionic components of the cell membrane and lead to cell death through various pore formations [20, 21]. However, this charge-induced binding mechanism also allows other anionic cell wall components such as LTA to absorb AMPs and protect the cell membrane from their destructive influence [22].

Other essential components of the bacterial cell membrane are lipoproteins, which are anchored via a diacylthioglycerol anchor in the cell membrane. Preprolipoproteins synthesized in the cytoplasma are retranslocated to the outer surface of the cytoplasmic membrane. They possess a consensus motif termed the lipobox, which is part of the N-terminal signal peptide cleavage site. The +1 cysteine residue of the lipobox undergoes lipid modification in a sequential process catalysed by three periplasmic enzymes to yield the mature lipoproteins. First, the preprolipoprotein diacylglyceryl transferase (Lgt) transfers a diacylglyerol moiety from phosphatidylglycerols to the sulfohydryl group of the +1 cysteine, thereby generating a thioester linkage [23]. Subsequently, the signal peptide is removed by the lipoprotein-specific signal peptidase (Lsp) [24‒26], leading to the intercalation of the diacylthioglycerol anchor into the cell membrane. For producing a mature triacylated lipoprotein, the lipoprotein N’N-acyl transferase (Lnt) catalyses aminoacylation of the α-amino group of the thiodiacylglyceryl cysteine [23]. The lipoprotein family is largely classified as substrate-binding proteins, which serve to sequester sugars, amino acids, small peptides, and essential metal ions to the cell via ATP-binding cassette transporter systems. Other members serve for resistance against antimicrobial substances, oxidative stress, maintenance of cell shape and osmotic stability, as well as protein folding or activation of various secreted cell surface proteins [9, 25, 27, 28]. Lipoproteins are involved in various stages during infection including, e.g., host cell adhesion and colonization, immune evasion, or survival in conquered host compartments [25, 29‒34]. Furthermore, lipoproteins activate the innate immune system via Toll-like receptor 2 (TLR2) and represent due to their conservation and immunogenicity promising candidates for a proteinaceous serotype-independent pneumococcal vaccine [35‒37].

Recently, we demonstrated that the lack of LTA in pneumococci had no impact on the morphology or growth even in minimal culture media, while being attenuated in pneumonia and sepsis mouse infection models [14]. Our data were corroborated when the CRISPR interference system was applied to identify pneumococcal genes pivotal under invasive in vivo conditions [38]. Due to discrepancy between in vitro and in vivo fitness of pneumococci lacking LTA, we speculated that already colonization might be affected and that despite invisible morphological changes LTA is essential for the integrity of the bacterial membrane. In the present study, we therefore assessed the impact of pneumococcal LTA on in vivo colonization using the non-invasive 19F serotype strain EF3030 [39]. We further tested whether also the tacL mutant of this strain is attenuated under invasive conditions and used the larvae of the greater wax moth, Galleria mellonella, as infection model. To understand the attenuation of pneumococci lacking LTA due to genetic ablation of TacL in more detail, we assessed in comparative studies alterations in the membrane fluidity, the abundance of lipoproteins, and susceptibility against AMPs by using our mutants and complemented strains generated in different pneumococcal serotypes.

Bacterial Strains and Growth Conditions

All strains used in this work are listed in Table 1. Escherichia coli strains were grown in liquid lysogeny broth medium (LB; Roth, Germany) or on LB agar plates (Roth, Germany) supplemented with erythromycin (2.5 μg/mL) or chloramphenicol (8 μg/mL). LB agar plates were incubated at 37°C and cultivation in liquid LB was performed at 37°C under agitation (120 rpm). S. pneumoniae serotype 2 strain D39 (NCTC7466), serotype 4 strain TIGR4, and serotype 19F strain EF3030, their isogenic mutants, and complemented strains were grown on Columbia blood agar plates (Thermo Scientific, Germany) supplemented with kanamycin (125 μg/mL), erythromycin (2.5 μg/mL), or chloramphenicol (8 μg/mL) if required. Liquid pneumococcal cultures were grown in Todd-Hewitt broth (THY; Roth, Germany) supplemented with 0.5% yeast extract and antibiotics if required. Blood agar plates were incubated at 37°C and 5% CO2 for 8–10 h. Cultivation in liquid cultures was performed at 37°C in a water bath without agitation.

Table 1.

List of bacterial strains used in this work

StrainsResistanceReference
1046 E. coli DH5α pUC18∆tacL AmpR, ErmR [14
1058 E. coli DH5α pBAV-tacL CmR [14
PN111 S. pneumoniae D39∆cps KmR [74
PN601 S. pneumoniae D39∆cpstacL KmR, ErmR [14
PN634 S. pneumoniae D39∆cpstacLtacL KmR, ErmR, CmR [14
PN259 S. pneumoniae TIGR4∆cps KmR [75
PN603 S. pneumoniae TIGR4∆cpstacL KmR, ErmR [14
PN636 S. pneumoniae TIGR4∆cpstacLtacL KmR, ErmR, CmR [14
SP408 S. pneumoniae EF3030 None [46
PN817 S. pneumoniae EF3030∆tacL ErmR This work 
PN818 S. pneumoniae EF3030∆tacLtacL ErmR, CmR This work 
StrainsResistanceReference
1046 E. coli DH5α pUC18∆tacL AmpR, ErmR [14
1058 E. coli DH5α pBAV-tacL CmR [14
PN111 S. pneumoniae D39∆cps KmR [74
PN601 S. pneumoniae D39∆cpstacL KmR, ErmR [14
PN634 S. pneumoniae D39∆cpstacLtacL KmR, ErmR, CmR [14
PN259 S. pneumoniae TIGR4∆cps KmR [75
PN603 S. pneumoniae TIGR4∆cpstacL KmR, ErmR [14
PN636 S. pneumoniae TIGR4∆cpstacLtacL KmR, ErmR, CmR [14
SP408 S. pneumoniae EF3030 None [46
PN817 S. pneumoniae EF3030∆tacL ErmR This work 
PN818 S. pneumoniae EF3030∆tacLtacL ErmR, CmR This work 

Construction of Pneumococcal Mutants and Complementation

The construction of the pneumococcal tacL-mutants in serotype 19F strain EF3030 was done using the recombinant plasmid pUC18∆tacL, which was constructed and described previously to generate tacL-mutants in D39/D39∆cps and TIGR4/TIGR4∆cps [14]. Transformation of EF3030 was performed as described previously [40]. EF3030∆tacL was complemented in trans using pBAV-tacL constructed as described [14]. Both, the mutant EF3030∆tacL and complemented strain EF3030∆tacLtacL were verified by PCR, sequencing (Eurofins Genomics, Germany), and qRT-PCR (online suppl. Fig. S1; for all online suppl. material, see https://doi.org/10.1159/000539934).

Oxidative Stress Assay

To analyse pneumococcal resistance to oxidative stress conditions, a hydrogen peroxide (H2O2)-stress assay was performed. Pneumococcal strains were grown until mid-exponential phase (A600 = 0.35–0.45) in THY medium. Each culture was split into 10 mL cultures and incubated with 0 mm, 5 mm, or 10 mm H2O2 for 30 min at 37°C. Afterwards, cultures were diluted in PBS (pH 7.4) and colony-forming units (CFUs) were determined by serial plating on blood agar plates (Thermo Scientific, Germany).

AMP Susceptibility Assay

To determine the susceptibility of pneumococcal wild-types and mutants to AMPs, LL-37 (InvivoGen, Germany) and CPL-1 (the endolysin was heterologously expressed in E. coli as described before by Hermoso et al. in [41]) were used. Pneumococcal strains were grown until mid-log phase (A600 = 0.35–0.45) in THY medium. Cultures were than split into 200 μL aliquots and incubated with 0 μm, 1 μm, or 2 μm LL-37 for 3 h at 37°C. Each sample was diluted in PBS (pH 7.4 for LL-37; pH 6.0 for CPL-1) and CFUs were determined by serial plating of relevant dilutions on Columbia blood agar plates (Thermo Scientific, Germany).

For treatment with CPL-1, pneumococcal strains were grown as described above and 0 μg/mL, 2 μg/mL (2.6 μm), or 50 μg/mL (65 μm) of the endolysin were added to the bacterial suspension. Pneumococcal lysis was determined by measuring absorption at 600 nm every 30 s.

Membrane Fluidity Staining

To visualize membrane regions of increased or decreased fluidity, the fluorescent membrane-integrating dye DiIC12 (1,1′-didodecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate; EnzoLife Sciences, Germany) was used. Pneumococcal strains were cultivated in THY medium containing 1 μg/mL DiIC12 (dissolved in DMSO) until mid-logarithmic phase (A600 = 0.35–0.45). Cultures were harvested at 3,275 × g and 30°C for 6 min and washed twice with pre-warmed THY containing 1% DMSO followed by washing with pre-warmed PBS (pH 7.4) containing 1% DMSO. A culture of each wild-type strain without dye was used as control. FACS Aria™ III (BD Biosciences, San Jose, CA, USA) was used for flow cytometric quantification of fluorescence [42]. Data analysis was carried out using FCS Express software (De Novo Software, Pasadena, CA, USA).

Generation of Antibodies

To analyse the abundance of pneumococcal lipoproteins on the surface of pneumococcal strains used in this study, polyclonal antibodies were generated in mice using routine immunization protocol as described previously [14]. Immunization of CD-1 mice was done by injecting 20 μg heterologously expressed non-lipidated proteins and Freund`s incomplete adjuvant (Sigma-Aldrich, Germany) (50:50 v/v) intraperitoneally. Mice were boosted with 20 μg protein and Freund`s incomplete adjuvants (Sigma-Aldrich, Germany) (50:50 v/v) after 14 and 28 days. After 48 days, mice were sacrificed and polyclonal IgGs were purified from serum using Protein A-Sepharose (Sigma-Aldrich, Germany).

Flow Cytometry

S. pneumoniae D39∆cps, TIGR4∆cps and EF3030 wild-type strains, isogenic tacL-mutants, and complemented strains were cultivated in THY medium until mid-logarithmic phase (A600 = 0.35–0.45). Bacteria were harvested at 3,275 × g for 6 min and washed 3 times with PBS (pH 7.4). Subsequently, strains were adjusted to the same A600 and 100 μL bacterial suspension was incubated with specific primary antibodies against various lipoproteins (1:500/1:250 in PBS) for 30–45 min at 37°C and 5% CO2 in 96-well U-bottom plates (Greiner Bio-One, Germany). Bacteria were washed 3 times and incubated with an Alexa-Fluor™ 488-labelled secondary goat-anti-mouse IgG antibody (Abcam, UK; 1:500 in PBS) for 30–45 min at 37°C and 5% CO2. Again, the samples were washed 3 times with PBS, and bacteria were then fixed with 1% para-formaldehyde (Roth, Germany) in PBS (pH 7.4) at 4°C overnight. Samples were subjected to flow cytometric analysis using a FACS Calibur™ (BD Biosciences, San Jose, CA, USA). The geometric mean fluorescence intensity was determined and illustrated as geometric mean fluorescence intensity multiplied with the percent of gated events. Data analysis was carried out using FCS Express software (De Novo Software, Pasadena, CA, USA).

Cell Culture Infection Experiment

Human nasopharyngeal epithelial cells (Detroit-562) were purchased from ATCC (ATCC CCL-138) and have been tested to be mycoplasma negative by PCR and scanning electron microscopy. Detroit-562 cells were used to analyse bacterial adherence to epithelial cells and for that purpose cultured in RPMI1640 medium (Capricorn Scientific, Germany) supplemented with 2% heat-inactivated fetal bovine serum (FBS; Sigma-Aldrich) and 1% sodium pyruvate (Cytiva, Germany) in 24-well plates (Greiner Bio-One, Germany). Two days prior to the infection with pneumococci, 1 × 105 cells were seeded per well. On the day of infection, 2 × 105 cells were washed with infection medium (RPMI1640 supplemented with 1% heat-inactivated FBS) and afterwards infected with 1 × 107 mid-exponentially grown pneumococci (multiplicity of infection of 50 bacteria per host cell) at 37°C and 5% CO2 in infection medium. After 4 h, non-adherent bacteria were removed by washing three times with PBS (pH 7.4). Subsequently, epithelial cells were treated with PBS  (pH 7.4) containing 5% saponin (Sigma-Aldrich, Germany) for 10 min at 37°C. The obtained suspension was diluted in PBS (pH 7.4) and total CFUs of adherent and invasive pneumococci were enumerated by serial plating on blood agar plates (Thermo Scientific, Germany).

Galleria mellonella Larvae Infection Model

Pneumococcal strains were cultured in THY medium until mid-exponential phase (A600 = 0.35–0.45) and washed with 0.9% sodium chloride. Subsequently, larvae of the greater wax moth, Galleria mellonella (Reptilienkosmos, Germany), with a weight of 0.3–0.4 g, were infected with 10 μL bacterial suspension (containing 1.0–5.0 × 105 bacteria) via an intrahemocelic route. A gastight microliter syringe (Hamilton, OH, USA) coupled with a repeating dispenser (Hamilton, OH, USA) was used to ensure equal infection doses. Each group of 10 larvae was stored at 37°C with sufficient food for 7 days post-infection and monitored daily. To determine the CFU, 10 larvae per group were anaesthetized with isoflurane (CP Pharma, Germany) and mechanically homogenized in 700 μL 0.9% sodium chloride using a T10 basic ULTRA-TURRAX (IKA, Germany). The bacterial suspension was serially diluted in 0.9% sodium chloride and plated on blood agar plates, which were incubated for 15 h at 37°C and 5% CO2. Food contained wheat bran, oatmeal, dry yeast, skim milk powder, honey, and glycerol in variable proportions.

Murine Nasopharyngeal Colonization Model

For the in vivo murine colonization studies, 8- to 10-week-old female CD-1 outbred mice (Janvier, France) were intranasally infected with serotype 19F strain EF3030, its isogenic tacL-mutant, and the complemented mutant. Strains were cultivated to mid-exponential phase (A600 = 0.35–0.45) in THY medium containing 10% heat-inactivated FBS. The infection dose of 20 μL was adjusted to 1.0–2.0 × 107 CFU in PBS  (pH 7.4) containing 1% heat-inactivated FBS. The CFU of the applied infection dose was verified by plating serial dilutions of the inoculum on blood agar plates. Mice were anaesthetized intraperitoneally with ketamine (Ketamin 10%; Selectavet, Germany) and xylazine (Xylasel; Selectavet, Germany) and intranasally infected with pneumococci as described previously [40]. At day 1, 3, 7, and 14 post-infection, nasopharyngeal and bronchoalveolar lavages were collected and the CFU was determined by plating serial dilutions on blood agar plates.

Statistical Analysis

Statistical significance between different groups was calculated using a one-way ANOVA (Kruskal-Wallis test) followed by Dunnett’s post-test for the mouse colonization model. Unpaired two-tailed Student’s t test (Mann-Whitney test) was performed to analyse the difference between two groups. Kaplan-Meier survival curves of mice were compared by the log-rank (Mantel-Cox) test. A p value of <0.05 was considered statistically significant. All statistical analyses were performed using GraphPad Prism version 5.0 (GraphPad Software, La Jolla, CA, USA). Statistically significant differences are indicated by *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.

LTAs Are Required to Cause Infections in the Galleria mellonella Model

The Galleria mellonella infection model has become increasingly important in recent years to investigate host-pathogen interactions under infection-related conditions. The larvae of the large wax moth G. mellonella, like other insects, do not possess an adaptive immune system [43]. However, they are producing a variety of AMPs and opsonins, which constitute a major part of the humoral host defence mechanisms. In this regard, the opsonin hemolin has been shown to bind lipopolysaccharides of Gram-negative and LTA of Gram-positive bacteria [44, 45]. As we have previously reported, the deficiency of TacL and consequently absence of LTA in the genetic background of the invasive serotype 2 strain D39 (D39ΔtacL) abrogates pneumonia and septicemia in murine infection models but does not affect in vitro growth [14]. Here, we infected G. mellonella larvae with a non-invasive pneumococcal strain, namely, serotype 19F strain EF3030 [46]. This strain colonizes the upper respiratory tract of mice after intranasal application for more than 14 days without inducing significant inflammatory responses or clinical symptoms [40, 47]. First, we aimed to elucidate whether 19F (EF3030) is harmful to G. mellonella and how the lack of LTA changes the virulence potential as well as the susceptibility to the humoral G. mellonella host defence mechanisms. Larvae were therefore infected with wild-type EF3030, the tacL-mutant EF3030ΔtacL and its complemented strain EF3030ΔtacLtacL. We observed that larvae infected with serotype 19F lacking LTA survived up to 7 days, which was significantly longer compared to larvae infected with the isogenic wild-type strain showing a survival for only 1 day. Similar to the wild-type, the complemented mutant EF3030ΔtacLtacL was severely harmful and killed larvae 1 day post-infection (Fig. 1). Interestingly, a difference in the amount of bacteria detected in the larvae could only be observed after 18 h. Animals infected with the mutant strain EF3030ΔtacL had a significantly lower number of bacteria compared to larvae infected with the wild-type EF3030 (Fig. 1d). These in vivo infection experiments indicate that serotype 19F is highly infectious in the G. mellonella model and that the loss of LTA results in a significantly higher susceptibility against the humoral response of the larvae.

Fig. 1.

Impact of LTA on Galleria mellonella infections with S. pneumoniae EF3030. a Infection process and survival of Galleria mellonella post-infection with S. pneumoniae EF3030 strains. Larvae were infected via the intrahemocelic route with 1.0–5.0 × 105 of EF3030 (wild-type), tacL-mutant EF3030ΔtacL, and complemented mutant EF3030ΔtacLtacL (n = 10). Survival was monitored for 7 days after infection. ****p < 0.0001 (log-rank [Mantel-Cox] test). b Virulence of S. pneumoniae EF3030 (wild-type), EF3030ΔtacL, and EF3030ΔtacLtacL in G. mellonella larvae. A time to death of ≥7 days indicates survival. Each data point represents one larvae. c Representative G. mellonella larvae infected with 1.0–5.0 × 105 bacteria of S. pneumoniae strains EF3030 (wild-type), EF3030ΔtacL, and EF3030ΔtacLtacL on day 1 post-infection. d Larvae were infected as in a and the bacterial load was determined after 3, 6, and 18 h. Thereafter, larvae were homogenized, samples serially diluted, and aliquots plated on blood agar plates to determine the CFU. The dotted line indicates limit of detection. *p < 0.05 (one-way ANOVA, Kruskal-wallis test followed by Dunnett’s post-test).

Fig. 1.

Impact of LTA on Galleria mellonella infections with S. pneumoniae EF3030. a Infection process and survival of Galleria mellonella post-infection with S. pneumoniae EF3030 strains. Larvae were infected via the intrahemocelic route with 1.0–5.0 × 105 of EF3030 (wild-type), tacL-mutant EF3030ΔtacL, and complemented mutant EF3030ΔtacLtacL (n = 10). Survival was monitored for 7 days after infection. ****p < 0.0001 (log-rank [Mantel-Cox] test). b Virulence of S. pneumoniae EF3030 (wild-type), EF3030ΔtacL, and EF3030ΔtacLtacL in G. mellonella larvae. A time to death of ≥7 days indicates survival. Each data point represents one larvae. c Representative G. mellonella larvae infected with 1.0–5.0 × 105 bacteria of S. pneumoniae strains EF3030 (wild-type), EF3030ΔtacL, and EF3030ΔtacLtacL on day 1 post-infection. d Larvae were infected as in a and the bacterial load was determined after 3, 6, and 18 h. Thereafter, larvae were homogenized, samples serially diluted, and aliquots plated on blood agar plates to determine the CFU. The dotted line indicates limit of detection. *p < 0.05 (one-way ANOVA, Kruskal-wallis test followed by Dunnett’s post-test).

Close modal

Impact of LTA on Pneumococcal Adherence and in vivo Colonization

Previously, we showed that S. pneumoniae strain D39ΔtacL lacking LTA had a lower ability to adhere to lung epithelial cells A549 [14]. Here, we were interested in the impact of LTA on pneumococcal colonization. However, strain D39 is unable to stably colonize the nasal tissue of mice. Because of its capability to colonize the nasopharynx and to be non-invasive [39], we therefore used serotype 19F strain EF3030 in the following in vitro adherence and in vivo colonization experiments. Prior to the mouse colonization experiments, we assessed the effect of LTA on adherence of serotype 19F strains to human nasopharyngeal epithelial cells (Detroit-562). The results showed a reduction of EF3030ΔtacL adherence to Detroit-562 epithelial cells, which is statistically significant. However, the overall impact on the pathophysiology is probably not high. The complemented mutant EF3030ΔtacLtacL showed no significant differences compared to the wild-type strain (Fig. 2a). Nevertheless, these data corroborate results we had reported previously for the isogenic tacL-mutant of the non-encapsulated strain D39Δcps when adherence was tested to A549 lung epithelial cells [14]. Altogether, our data verify the role of LTA for pneumococcal attachment to epithelial cells.

Fig. 2.

Pneumococcal LTA is required for in vitro adherence and in vivo colonization. a Effect of LTA on pneumococcal adherence to nasopharyngeal epithelial cells. Human nasopharyngeal epithelial cells (Detroit-562) were infected with S. pneumoniae serotype 19F strain EF3030, its isogenic tacL-mutant (EF3030ΔtacL), or complemented mutant (EF3030∆tacLtacL) using an MOI of 50. After 4 h, epithelial cells were washed, lysed and the resulting suspensions were plated on Columbia blood agar plates to quantify the number of colony-forming units (CFUs). All values are represented as CFU/mL and expressed as mean ± SD (n ≥ 3). **p < 0.005 (Student’s unpaired t test). b Impact of pneumococcal LTA on colonization. 8- to 10-weeks old female CD-1 mice (n = 7) were intranasally infected with EF3030, the mutant EF3030∆tacL, or the complemented strain EF3030∆tacLtacL. Nasopharyngeal and broncheoalveolar lavages were plated on Colombia blood agar plates to quantify bacterial load after 1, 3, 7, and 14 days post-infection. Results are shown as scatter plots. Each dot represents one individual mouse (n = 7). The dotted line indicates limit of detection. *p < 0.05 (one-way ANOVA, Kruskal-wallis test followed by Dunnett’s post-test).

Fig. 2.

Pneumococcal LTA is required for in vitro adherence and in vivo colonization. a Effect of LTA on pneumococcal adherence to nasopharyngeal epithelial cells. Human nasopharyngeal epithelial cells (Detroit-562) were infected with S. pneumoniae serotype 19F strain EF3030, its isogenic tacL-mutant (EF3030ΔtacL), or complemented mutant (EF3030∆tacLtacL) using an MOI of 50. After 4 h, epithelial cells were washed, lysed and the resulting suspensions were plated on Columbia blood agar plates to quantify the number of colony-forming units (CFUs). All values are represented as CFU/mL and expressed as mean ± SD (n ≥ 3). **p < 0.005 (Student’s unpaired t test). b Impact of pneumococcal LTA on colonization. 8- to 10-weeks old female CD-1 mice (n = 7) were intranasally infected with EF3030, the mutant EF3030∆tacL, or the complemented strain EF3030∆tacLtacL. Nasopharyngeal and broncheoalveolar lavages were plated on Colombia blood agar plates to quantify bacterial load after 1, 3, 7, and 14 days post-infection. Results are shown as scatter plots. Each dot represents one individual mouse (n = 7). The dotted line indicates limit of detection. *p < 0.05 (one-way ANOVA, Kruskal-wallis test followed by Dunnett’s post-test).

Close modal

To confirm our in vitro findings in vivo, we applied a mouse colonization model. Groups of CD-1 outbred mice were intranasally infected with wild-type serotype 19F strain EF3030, its isogenic tacL-mutant, and the in trans complemented mutant. The wild-type and complemented mutant EF3030∆tacL∇tacL showed a stable colonization of the nasopharynx for 14 days (Fig. 2b). In contrast, mice infected with the mutant strain lacking LTA had lower numbers of bacteria in the nasopharynx. This difference was most prominent at day 14 post-infection, while unexpectedly no differences were shown at day 7 post-infection. Similar to our earlier reported data [47], 19F bacteria, irrespectively whether wild-type or tacL-mutant, were isolated only at low numbers from the bronchoalveolar lavage and if at all only at earlier time points post-infection. Strikingly, mice infected with the complemented mutant EF3030∆tacL∇ tacL showed a relatively stable number of bacteria in the bronchoalveolar lavage up to 7 days post-infection (Fig. 2b). In the in trans complemented mutant, the tacL gene is located on a plasmid and expressed from a strong promotor [14]. Taken together, our results clearly show the importance of LTA for pneumococcal adherence and colonization under in vivo conditions.

Lack of LTAs Perturbs Membrane Composition and Fluidity

Previously, we reported that the abundance of CBPs on the pneumococcal surface has not changed substantially in the absence of LTA. The absence of any impact on CBPs is probably due to the 9:1 ratio of WTA/LTA, meaning that 90% of the phosphorylcholine-binding sites are still available for CBPs in pneumococci lacking LTA due to genetic ablation of tacL [14, 48]. Because LTA is intercalated in the cell membrane via a glycosyldiacylglycerol anchor, we hypothesized that the absence of LTA might change the membrane properties and influence the membrane composition, although the glycosylglycerolipids themselves are still present. We therefore investigated at first the surface abundance of pneumococcal prototype lipoproteins, which are intercalated in the cell membrane by their diacylthioglycerol anchor [23]. The results of our flow cytometric analysis showed for serotype 2 strain D39∆cps and serotype 19F strain EF3030 a significantly higher abundance of surface-exposed lipoproteins PsaA, SlrA, and DacB in the tacL-mutants compared to the isogenic wild-type strains (Fig. 3a). In contrast, the complemented mutants showed amounts of lipoproteins comparable to the corresponding wild-type strain. In a complementary approach, we also observed changes in lipoprotein levels for PsaA, SlrA, and DacB, when whole bacterial cell lysates of strain D39∆cps and its isogenic mutant as well as the complemented strain were used for quantitative Western blot analysis (online suppl. Fig. S2). Remarkably, the effect of a higher lipoprotein abundance in the tacL-mutant is less pronounced for TIGR4∆cpstacL.

Fig. 3.

LTA affects abundance of pneumococcal lipoproteins and membrane stiffness. a Abundance of lipoproteins PsaA, SlrA, and DacB on the pneumococcal cell surface. Bacteria were grown in THY medium to A600 = 0.35–0.45, washed in PBS (pH 7.4), and incubated with lipoprotein-specific mouse polyclonal IgG generated against non-lipidated pneumococcal lipoproteins PsaA, SlrA, and DacB, respectively [29, 31]. For the detection of lipoproteins by flow cytometry, an Alexa-Fluor™ 488-labelled secondary goat-anti-mouse IgG antibody was used. The relative quantity of lipoproteins on the surface of pneumococci is shown as a box-whisker graph. b Influence of LTA on the stiffness of the pneumococcal membrane. To measure the membrane fluidity of pneumococci expressing or lacking LTA, bacteria were cultured in THY medium containing 1 μg/mL DiIC12 dye to A600 = 0.35–0.45. After washing the sedimented bacteria with pre-warmed THY and PBS (pH 7.4) containing 1% DMSO, the fluorescence of pneumococci was measured by flow cytometry. All values are represented as the geometrical mean fluorescence intensity multiplied with percent-gated events and are expressed as mean ± SD (n ≥ 3). *p < 0.05; **p < 0.005; ***p < 0.0005; ****p < 0.0001 (Student’s unpaired t test). c Stacked histograms of one replicate for each strain, showing the cell count over the fluorescence intensity.

Fig. 3.

LTA affects abundance of pneumococcal lipoproteins and membrane stiffness. a Abundance of lipoproteins PsaA, SlrA, and DacB on the pneumococcal cell surface. Bacteria were grown in THY medium to A600 = 0.35–0.45, washed in PBS (pH 7.4), and incubated with lipoprotein-specific mouse polyclonal IgG generated against non-lipidated pneumococcal lipoproteins PsaA, SlrA, and DacB, respectively [29, 31]. For the detection of lipoproteins by flow cytometry, an Alexa-Fluor™ 488-labelled secondary goat-anti-mouse IgG antibody was used. The relative quantity of lipoproteins on the surface of pneumococci is shown as a box-whisker graph. b Influence of LTA on the stiffness of the pneumococcal membrane. To measure the membrane fluidity of pneumococci expressing or lacking LTA, bacteria were cultured in THY medium containing 1 μg/mL DiIC12 dye to A600 = 0.35–0.45. After washing the sedimented bacteria with pre-warmed THY and PBS (pH 7.4) containing 1% DMSO, the fluorescence of pneumococci was measured by flow cytometry. All values are represented as the geometrical mean fluorescence intensity multiplied with percent-gated events and are expressed as mean ± SD (n ≥ 3). *p < 0.05; **p < 0.005; ***p < 0.0005; ****p < 0.0001 (Student’s unpaired t test). c Stacked histograms of one replicate for each strain, showing the cell count over the fluorescence intensity.

Close modal

These results intrigued us to investigate the pneumococcal membrane fluidity using the fluorescence dye DiIC12. This dye shows a high affinity to membrane parts having an increased membrane fluidity [49]. After incubation of our set of pneumococcal strains with DiIC12, we quantified the bacterial fluorescence intensity by flow cytometry. We observed a significant lower amount of DiIC12 dye incorporated into the bacterial cell membrane of tacL-mutants when compared to isogenic wild-type strains D39∆cps serotype 2 and EF3030 serotype 19F. These data suggest that the membrane fluidity in pneumococci lacking LTA is decreased. Importantly, the complemented strains of the above-mentioned serotypes showed significantly higher amount of DiIC12 dye incorporated into the cell membrane when compared to the wild-type. This effect is, as mentioned, most likely due to the higher TacL expression in the in trans complemented strains, thereby showing the gene dosage effect (Fig. 3b, c). Serotype 4 strain TIGR4 showed, similar to our findings for the lipoprotein abundancies, neither lower DiIC12 levels in the tacL-mutant nor higher DiIC12 incorporation in the complemented mutant. Taken together, changes in lipoprotein abundance and lower DiIC12 dye incorporation in the bacterial cell membrane of LTA-deficient pneumococci D39∆cpstacL and EF3030∆tacL suggest that the fluidity of the cell membrane is reduced in these strains and a stiffer membrane is disadvantageous for pneumococci under in vivo conditions.

LTA Absence Leads to a Higher Susceptibility to Host Innate Immune Defence Mechanisms

The integrity of the PGN and cytoplasmic membrane surrounding the cytoplasm of Gram-positive bacteria is crucial for cellular homeostasis, to survive environmental stress conditions, and to resist innate immune defence mechanisms [50‒52]. To understand the impact of the altered membrane composition in the absence of LTA, we compared the stress resistance of wild-type strains and tacL-mutants. In the presence of 5 mm and 10 mm hydrogen peroxide (H2O2), which causes oxidative stress, we observed that the presence of LTA is essential for resistance against oxidative stress. Regardless of the genetic background and serotype tested, tacL-mutants lacking LTA showed a significantly lower survival rate compared to the isogenic wild-type strain possessing LTA (Fig. 4a). The complemented strains showed a behaviour similar to the wild-type D39∆cps, EF3030, and TIGR4∆cps, respectively (Fig. 4a).

Fig. 4.

Susceptibility of pneumococcal cells to oxidative stress and AMPs. All pneumococcal strains were cultured in THY medium until reaching mid-exponential growth phase. a Strains were then split and incubated with indicated concentrations of hydrogen peroxide (0 mm, 5 mm, or 10 mm). Colony-forming units (CFUs) were enumerated by plating out dilution series on blood agar plates and survival was calculated. CFU determined for pneumococci strains in the absence of LL-37 was set to 100%. b All strains were split up and incubated with indicated concentrations of LL-37 (0 μm, 1 μm, or 2 μm). CFUs were enumerated by plating out dilution series on blood agar plates and survival was calculated. CFU determined for pneumococci strains in the absence of LL-37 was set to 100%. c Pneumococcal strains were spiked with the indicated concentration of CPL-1 (0 μg/mL, 2 μg/mL, or 50 μg/mL). Subsequently, the decrease in absorbance at 600 nm was measured every 30 s for 5 min to determine cell lysis. All values are expressed as means ± SD (n ≥ 3). *p < 0.05; **p < 0.005; ***p < 0.0005; ****p < 0.0001 (Student’s unpaired t test).

Fig. 4.

Susceptibility of pneumococcal cells to oxidative stress and AMPs. All pneumococcal strains were cultured in THY medium until reaching mid-exponential growth phase. a Strains were then split and incubated with indicated concentrations of hydrogen peroxide (0 mm, 5 mm, or 10 mm). Colony-forming units (CFUs) were enumerated by plating out dilution series on blood agar plates and survival was calculated. CFU determined for pneumococci strains in the absence of LL-37 was set to 100%. b All strains were split up and incubated with indicated concentrations of LL-37 (0 μm, 1 μm, or 2 μm). CFUs were enumerated by plating out dilution series on blood agar plates and survival was calculated. CFU determined for pneumococci strains in the absence of LL-37 was set to 100%. c Pneumococcal strains were spiked with the indicated concentration of CPL-1 (0 μg/mL, 2 μg/mL, or 50 μg/mL). Subsequently, the decrease in absorbance at 600 nm was measured every 30 s for 5 min to determine cell lysis. All values are expressed as means ± SD (n ≥ 3). *p < 0.05; **p < 0.005; ***p < 0.0005; ****p < 0.0001 (Student’s unpaired t test).

Close modal

Cathelicidin, also known as LL-37, is a cationic AMP (cAMP) produced by epithelial and circulating immune cells, representing the most prominent cAMP in bronchoalveolar fluids [53]. To assess the role of LTA in cAMP resistance, pneumococci were cultured in the presence or absence of different LL-37 concentrations (0 μm, 1 μm, or 2 μm) and the survival rate was determined. Our data suggest that pneumococcal resistance to LL-37 correlates with the presence of surface-exposed LTA, which is shown by lower survival rate of the tacL-mutants (Fig. 4b). However, statistical significance could only be shown for serotype 2 strain D39∆cps, while serotype 19F strain EF3030 showed only a trend towards a higher susceptibility against LL-37 in the absence of LTA (Fig. 4b).

In addition, we tested the role of LTA for the susceptibility against the Cp-1 phage-encoded lysozyme CPL-1. This enzyme contains a specific choline-binding domain and acts as a muramidase. Therefore, it specifically binds to the phosphorylcholine decorations of the pneumococcal TAs and induces lysis of the bacterial cell wall [54]. Due to the high potency, we tested pneumococcal survival in the presence of a low (2 μg/mL) and high (50 μg/mL) lysozyme concentration. For serotype 4 wild-type strain TIGR4 and its tacL-mutant, we could not measure that loss of LTA had an impact on pneumococcal lysis, independent of the used CPL-1 concentration (Fig. 4c). In contrast, serotype 2 and 19F strains lacking LTA showed a significantly higher susceptibility to CPL-1 than the isogenic wild-type strains (Fig. 4c). Remarkably, complemented tacL-mutants of 19F and D39∆cps showed even a slightly higher resistance to CPL-1. Again, this is most likely a gene dosage effect and caused by the higher numbers of tacL-gene copies in these strains (online suppl. Fig. S1). Taken together, the absence of LTA leads to a higher susceptibility to oxidative stress, which is most likely not influenced by the observed, serotype-dependent changes in membrane fluidity because all three analysed serotypes showed similar changes. For the analysed susceptibility to AMPs, no or only very minor differences were detected. However, the tested mutant strains of serotype 2 and 19F showed an overall similar trend in their susceptibility, while this effect was not observed for the serotype 4 strain TIGR4 (Fig. 4b, c).

LTA plays an important role in the network of different polymers that form the bacterial cell wall. Due to their ability to bind CBPs via their non-covalent interaction with phosphorylcholine residues of TAs, these polysaccharides are important for cell wall remodelling and thus for the pneumococcal cell physiology. In some Gram-positive bacteria, e.g., in Staphylococcus aureus, the lack of LTA leads to lethal cell defects due to dramatic alterations of the bacterial cell morphology [55, 56]. For S. pneumoniae, we have recently shown that the genetic ablation of the TA ligase TacL leads to a complete loss of LTA. Unexpectedly, the bacterial cells showed a normal growth behaviour. Nevertheless, the invasive serotype 2 strain D39 lacking LTA (D39ΔtacL) showed a reduced ability to adhere to lung epithelial cells and was significantly attenuated in a mouse pneumonia and sepsis infection model [14]. Here, we demonstrate that the reduction in virulence is not only relevant for invasive serotypes but also for non-invasive serotypes such as S. pneumoniae serotype 19F strain EF3030. This strain was isolated from a patient suffering on otitis media and stably colonizes the respiratory tract of mice without causing any clinical symptoms [46, 47]. EF3030 lacking LTA showed a slightly reduced ability to adhere to human nasopharyngeal epithelial cells in our in vitro infection model. In accordance with these results that most likely explain our in vivo data, EF3030 lacking TacL and hence LTA showed a minor reduction in bacterial load in the nasopharynx of the murine colonization model over the course of 14 days. Interestingly, the wild-type strain EF3030, which is non-invasive in mice, showed high virulence in the G. mellonella larval infection model after intrahemocelic injection of the bacteria. EF3030 lacking LTA was significantly attenuated in the G. mellonella model, while the complemented mutant was as virulent as the wild-type. Because of their humoral system that is − like that of most other insects − based on a complex system of opsonins, AMPs, and lysozymes, our data implied that these substances in particular show a higher efficiency against LTA-deficient strains [44].

In general, it is not well understood how LTA compositional variation modulates the lipid membrane stability and integrity. Liposomes mimicking the lipid composition of the Gram-positive model organism Bacillus subtilis have been used to investigate structural changes in the lipid layer when the amount of LTA is changed. The study clearly indicated that variations in LTA affect the stability and rigidity of the phospholipid membrane [57]. Taking these findings and our data into account, we asked whether lack of LTA in pneumococcal strains also affects the membrane stability. Indeed, our data showed a reduction in membrane fluidity in the absence of LTA, which resulted in a lower incorporation of the fluorescent membrane dye DiIC12. The lower membrane fluidity is probably due to a variation in the protein/lipid ratio. Studies on membrane composition showed that the membrane flexibility is negatively affected when the ratio shifts to the protein sides [58]. Proteins directly intercalated into the membrane are the lipoproteins. Comparing the amount of lipoproteins between the wild-type strains and their respective isogenic mutants indicated a higher lipoprotein abundance on the surface of D39∆cps and EF3030 lacking LTA when using DacB, PsaA, and SlrA as prototypes. However, for TIGR4 ∆cps only PsaA was significantly enhanced in the tacL-mutant, while DacB and SlrA were similarly present as in wild-type. The TIGR4 data are in accordance with our flow cytometric analysis using the membrane dye since we did not measure differences in membrane flexibility among TIGR4∆cps strains. These data suggest that the effects induced in the absence of LTA might be strain specific, a phenomenon that has to be investigated further.

Various studies have shown that the membrane composition of bacteria changes in response to environmental influences [59]. We therefore investigated the susceptibility against oxidative stress using H2O2. All tacL-mutants including the one from TIGR4 showed a high susceptibility against oxidative stress induced by H2O2. Even the higher abundance of surface-exposed PsaA, which is the substrate-binding protein of the manganese ATP-binding cassette transporter and has been reported to be important for oxidative stress resistance stress [60], had no protective effect. The high susceptibility of TacL-deficient pneumococci to H2O2 treatment is in a hitherto unknown manner linked to the reduced membrane fluidity and lack of LTA, because the increased susceptibility was also observed for TIGR4 strains, which showed no changes in membrane fluidity. However, the increased resistance observed for the complemented mutants and in particular in strain EF3030∆tacLtacL might suggest that the LTA is directly or indirectly influencing the defence against oxidative stress. The higher tacL expression in the complemented strain leads most likely to an increased incorporation of LTA into the cell membrane, which would also support the results of the membrane fluidity analyses. A recent study showed that PspA-mediated aggregation protects pneumococcal desiccation, when incubated on abiotic surfaces [61]. To elucidate the impact of LTA on desiccation, we incubated EF3030, its isogenic tacL-mutant, and complemented strain on glass slides at room temperature. Under the selected conditions and in the absence of LTA, pneumococcal killing is accelerated (online suppl. Fig. S3). This might be due to a lower resistance against osmotic stress, which is caused by higher salt concentration during desiccation. Thus, the H2O2 results are corroborated.

To test the susceptibility against AMPs, we incubated the investigated strains with human cathelicidin LL-37, which is primarily produced by epithelial cells and could therefore also be detected in the nasopharynx and in the human lung fluid [53, 62‒64]. IL-37 plays a role not only as a defence mechanism against invasive bacteria but also against colonizing bacteria. Like other AMPs, it acts by attaching to the negatively charged bacterial membrane and disrupting it by forming transmembrane pores [65]. Our results indicated a significantly increased susceptibility of D39∆cps lacking LTA against LL-37, while EF3030ΔtacL showed a trend towards higher susceptibility compared to the wild-type and TIGR4∆cpsΔtacL was similar susceptible to its wild-type strain. The higher in vitro susceptibility of strains lacking LTA to this AMP may also play a role in invasive infections or during in vivo colonization. One defence mechanism of Gram-positive bacteria against cAMPs is the D-alanylation of TAs. The incorporation of these positively charged residues leads to a charge-dependent defence against cAMPs. The general capability of some S. pneumoniae strains to decorate their LTA with alanine has been shown; however, the degree seems to be different among serotypes [12, 66]. Interestingly, proteomic analyses of S. pneumoniae have identified 52 protein candidates that may play an important role in resistance to LL-37, with the TA flippase TacF being one of these proteins [67, 68].

Our previous study have already shown that although the deletion of TacL leads to a total loss of LTA, the amount of P-Cho residues detected in the cell wall remains unchanged [14]. This could be due to the fact that the majority of the TAs are represented by the WTA or because the existing TA precursor chains are increasingly incorporated as WTA in TacL-deficient strains. The higher susceptibility against the CPL-1 lysozyme in serotype 2 strain D39 and serotype 19F strain EF3030 is therefore most likely not due to lower CPL-1 binding sites. In addition, the total amount of CBPs seems to be unaltered [14]. CBPs such as the autolysins LytA, LytB, or LytC are involved in the remodelling of the PGN or, like Pce (phosphorylcholine esterase), in the decoration of P-Cho residues to the TAs [69‒73]. However, when CPL-1 is exogenously added and bound to available P-Cho residues not decorated with CBPs, the combination of a lower membrane flexibility and additional enzyme cleaving PGN might result in a higher killing of S. pneumoniae lacking LTA. The different susceptibility measured for the non-encapsulated D39 with a sensitivity already at 2 μg/mL and encapsulated EF3030 with a sensitivity at 50 μg/mL might be due to their capsule phenotype. The capsule in EF3030, although not very thick, may mask P-Cho binding sites for CPL-1.

In conclusion, this study indicates the critical role of LTA during pneumococcal colonization and in escaping the innate immune defence mechanisms of the host. Our data showed that alterations in LTA are associated with changes in membrane flexibility and integrity and the balanced expression of WTA and LTA is required to maintain the cell wall integrity and thus resistance against host defence mechanisms.

We would like to thank Professor Anders Håkansson (Lund University, Sweden) for providing S. pneumoniae 19F strain EF3030. We thank Kristine Sievert-Giermann and Peggy Stremlow (Department of Molecular Genetics and Infection Biology, University of Greifswald) for technical assistance.

The animal experiments were conducted in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals (National Research Council, Washington, DC, USA), the guidelines of the Ethics Committee at the University of Greifswald, and the German regulations of the Society for Laboratory Animal Science (GVSOLAS) and the European Health Law of the Federation of Laboratory Animal Science Associations (FELASA). The study protocol was reviewed and approved by Landesamt für Landwirtschaft, Lebensmittelsicherheit und Fischerei Mecklenburg-Vorpommern (LALLFV M-V, Rostock, Germany) and the LALLFV M-V ethical board (LALLF M-V permit No. 7221.3-1-006/23). All efforts were made to minimize suffering, ensure the highest ethical standard, and adhere to the 3R principle (reduction, refinement, and replacement).

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

This work was supported by grants of the Deutsche Forschungsgemeinschaft to S.H. (HA 3125/5-2) and N.G. (GI 979/1-2). The funders had no role in study design, decision to publish, or manuscript preparation.

The experiments were conceived and designed by M.B. and S.H. Experiments were performed by M.B., A.P., and T.P.K. Mutant was constructed by M.B. and J.V.N. Editorial advice was given by S.H., T.P.K., and N.G. Data analysis was done by and manuscript was written by M.B. Revision was done by N.G. and S.H. All authors contributed to the article and approved the submitted version.

All relevant data are contained within the manuscript and supporting information files.

1.
GBD 2016 Lower Respiratory Infections Collaborators
.
Estimates of the global, regional, and national morbidity, mortality, and aetiologies of lower respiratory infections in 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016
.
Lancet Infect Dis
.
2018
;
18
(
11
):
1191
210
.
2.
Orihuela
C
,
Brown
J
,
Hammerschmidt
S
, editors.
Streptococcus pneumoniae: Molecular mechanisms of host-pathogen interactions
.
Amsterdam
:
Academic Press
;
2015
.
3.
Hyams
C
,
Camberlein
E
,
Cohen
JM
,
Bax
K
,
Brown
JS
.
The Streptococcus pneumoniae capsule inhibits complement activity and neutrophil phagocytosis by multiple mechanisms
.
Infect Immun
.
2010
;
78
(
2
):
704
15
.
4.
Calix
JJ
,
Porambo
RJ
,
Brady
AM
,
Larson
TR
,
Yother
J
,
Abeygunwardana
C
, et al
.
Biochemical, genetic, and serological characterization of two capsule subtypes among Streptococcus pneumoniae serotype 20 strains: discovery of a new pneumococcal serotype
.
J Biol Chem
.
2012
;
287
(
33
):
27885
94
.
5.
Ganaie
F
,
Maruhn
K
,
Li
C
,
Porambo
RJ
,
Elverdal
PL
,
Abeygunwardana
C
, et al
.
Structural, genetic, and serological elucidation of Streptococcus pneumoniae serogroup 24 serotypes: discovery of a new serotype, 24C, with a variable capsule structure
.
J Clin Microbiol
.
2021
;
59
(
7
):
e0054021
.
6.
Ganaie
F
,
Saad
JS
,
McGee
L
,
van Tonder
AJ
,
Bentley
SD
,
Lo
SW
, et al
.
A new pneumococcal capsule type, 10D, is the 100th serotype and has a large cps fragment from an oral Streptococcus
.
mBio
.
2020
;
11
(
3
):
e00937-20
.
7.
Ganaie
FA
,
Saad
JS
,
Lo
SW
,
McGee
L
,
Bentley
SD
,
van Tonder
AJ
, et al
.
Discovery and characterization of pneumococcal serogroup 36 capsule subtypes, serotypes 36A and 36B
.
J Clin Microbiol
.
2023
;
61
(
4
):
e0002423
.
8.
Geno
KA
,
Gilbert
GL
,
Song
JY
,
Skovsted
IC
,
Klugman
KP
,
Jones
C
, et al
.
Pneumococcal capsules and their types: past, present, and future
.
Clin Microbiol Rev
.
2015
;
28
(
3
):
871
99
.
9.
Bergmann
S
,
Hammerschmidt
S
.
Versatility of pneumococcal surface proteins
.
Microbiology (Reading)
.
2006
;
152
(
Pt 2
):
295
303
.
10.
Gamez
G
,
Hammerschmidt
S
.
Combat pneumococcal infections: adhesins as candidates for protein-based vaccine development
.
Curr Drug Targets
.
2012
;
13
(
3
):
323
37
.
11.
Voss
S
,
Gámez
G
,
Hammerschmidt
S
.
Impact of pneumococcal microbial surface components recognizing adhesive matrix molecules on colonization
.
Mol Oral Microbiol
.
2012
;
27
(
4
):
246
56
.
12.
Gisch
N
,
Kohler
T
,
Ulmer
AJ
,
Müthing
J
,
Pribyl
T
,
Fischer
K
, et al
.
Structural reevaluation of Streptococcus pneumoniae lipoteichoic acid and new insights into its immunostimulatory potency
.
J Biol Chem
.
2013
;
288
(
22
):
15654
67
.
13.
Denapaite
D
,
Brückner
R
,
Hakenbeck
R
,
Vollmer
W
.
Biosynthesis of teichoic acids in Streptococcus pneumoniae and closely related species: lessons from genomes
.
Microb Drug Resist
.
2012
;
18
(
3
):
344
58
.
14.
Heß
N
,
Waldow
F
,
Kohler
TP
,
Rohde
M
,
Kreikemeyer
B
,
Gómez-Mejia
A
, et al
.
Lipoteichoic acid deficiency permits normal growth but impairs virulence of Streptococcus pneumoniae
.
Nat Commun
.
2017
;
8
(
1
):
2093
.
15.
Vollmer
W
,
Massidda
O
,
Tomasz
A
.
The cell wall of Streptococcus pneumoniae
.
Microbiol Spectr
.
2019
;
7
(
3
).
16.
Knaack
W
,
Hölzl
G
,
Gisch
N
.
Structural analysis of glycosylglycerolipids using NMR spectroscopy
.
Methods Mol Biol
.
2021
;
2295
:
249
72
.
17.
Meiers
M
,
Volz
C
,
Eisel
J
,
Maurer
P
,
Henrich
B
,
Hakenbeck
R
.
Altered lipid composition in Streptococcus pneumoniae cpoA mutants
.
BMC Microbiol
.
2014
;
14
:
12
.
18.
Trombe
MC
,
Lanéelle
MA
,
Lanéelle
G
.
Lipid composition of aminopterin-resistant and sensitive strains of Streptococcus pneumoniae. Effect of aminopterin inhibition
.
Biochim Biophys Acta
.
1979
;
574
(
2
):
290
300
.
19.
Joyce
LR
,
Guan
Z
,
Palmer
KL
.
Streptococcus pneumoniae, S. pyogenes and S. agalactiae membrane phospholipid remodelling in response to human serum
.
Microbiology (Reading)
.
2021
;
167
(
5
):
001048
.
20.
He
K
,
Ludtke
SJ
,
Worcester
DL
,
Huang
HW
.
Neutron scattering in the plane of membranes: structure of alamethicin pores
.
Biophys J
.
1996
;
70
(
6
):
2659
66
.
21.
Pieta
P
,
Mirza
J
,
Lipkowski
J
.
Direct visualization of the alamethicin pore formed in a planar phospholipid matrix
.
Proc Natl Acad Sci U S A
.
2012
;
109
(
52
):
21223
7
.
22.
Scott
MG
,
Gold
MR
,
Hancock
REW
.
Interaction of cationic peptides with lipoteichoic acid and Gram-positive bacteria
.
Infect Immun
.
1999
;
67
(
12
):
6445
53
.
23.
Goolab
S
,
Roth
RL
,
van Heerden
H
,
Crampton
MC
.
Analyzing the molecular mechanism of lipoprotein localization in Brucella
.
Front Microbiol
.
2015
;
6
:
1189
.
24.
Kovacs-Simon
A
,
Titball
RW
,
Michell
SL
.
Lipoproteins of bacterial pathogens
.
Infect Immun
.
2011
;
79
(
2
):
548
61
.
25.
Kohler
S
,
Voß
F
,
Gómez Mejia
A
,
Brown
JS
,
Hammerschmidt
S
.
Pneumococcal lipoproteins involved in bacterial fitness, virulence, and immune evasion
.
FEBS Lett
.
2016
;
590
(
21
):
3820
39
.
26.
Pribyl
T
,
Moche
M
,
Dreisbach
A
,
Bijlsma
JJE
,
Saleh
M
,
Abdullah
MR
, et al
.
Influence of impaired lipoprotein biogenesis on surface and exoproteome of Streptococcus pneumoniae
.
J Proteome Res
.
2014
;
13
(
2
):
650
67
.
27.
Pérez-Dorado
I
,
Galan-Bartual
S
,
Hermoso
JA
.
Pneumococcal surface proteins: when the whole is greater than the sum of its parts
.
Mol Oral Microbiol
.
2012
;
27
(
4
):
221
45
.
28.
Alloing
G
,
de Philip
P
,
Claverys
JP
.
Three highly homologous membrane-bound lipoproteins participate in oligopeptide transport by the Ami system of the Gram-positive Streptococcus pneumoniae
.
J Mol Biol
.
1994
;
241
(
1
):
44
58
.
29.
Abdullah
MR
,
Gutiérrez-Fernández
J
,
Pribyl
T
,
Gisch
N
,
Saleh
M
,
Rohde
M
, et al
.
Structure of the pneumococcal l,d-carboxypeptidase DacB and pathophysiological effects of disabled cell wall hydrolases DacA and DacB
.
Mol Microbiol
.
2014
;
93
(
6
):
1183
206
.
30.
Cron
LE
,
Bootsma
HJ
,
Noske
N
,
Burghout
P
,
Hammerschmidt
S
,
Hermans
PWM
.
Surface-associated lipoprotein PpmA of Streptococcus pneumoniae is involved in colonization in a strain-specific manner
.
Microbiology (Reading)
.
2009
;
155
(
Pt 7
):
2401
10
.
31.
Hermans
PWM
,
Adrian
PV
,
Albert
C
,
Estevão
S
,
Hoogenboezem
T
,
Luijendijk
IHT
, et al
.
The streptococcal lipoprotein rotamase A (SlrA) is a functional peptidyl-prolyl isomerase involved in pneumococcal colonization
.
J Biol Chem
.
2006
;
281
(
2
):
968
76
.
32.
Rajam
G
,
Anderton
JM
,
Carlone
GM
,
Sampson
JS
,
Ades
EW
.
Pneumococcal surface adhesin A (PsaA): a review
.
Crit Rev Microbiol
.
2008
;
34
(
3–4
):
163
73
.
33.
Saleh
M
,
Bartual
SG
,
Abdullah
MR
,
Jensch
I
,
Asmat
TM
,
Petruschka
L
, et al
.
Molecular architecture of Streptococcus pneumoniae surface thioredoxin-fold lipoproteins crucial for extracellular oxidative stress resistance and maintenance of virulence
.
EMBO Mol Med
.
2013
;
5
(
12
):
1852
70
.
34.
Schmidt
F
,
Kakar
N
,
Meyer
TC
,
Depke
M
,
Masouris
I
,
Burchhardt
G
, et al
.
In vivo proteomics identifies the competence regulon and AliB oligopeptide transporter as pathogenic factors in pneumococcal meningitis
.
Plos Pathog
.
2019
;
15
(
7
):
e1007987
.
35.
Moffitt
K
,
Skoberne
M
,
Howard
A
,
Gavrilescu
LC
,
Gierahn
T
,
Munzer
S
, et al
.
Toll-like receptor 2-dependent protection against pneumococcal carriage by immunization with lipidated pneumococcal proteins
.
Infect Immun
.
2014
;
82
(
5
):
2079
86
.
36.
Voß
F
,
Kohler
TP
,
Meyer
T
,
Abdullah
MR
,
van Opzeeland
FJ
,
Saleh
M
, et al
.
Intranasal vaccination with lipoproteins confers protection against pneumococcal colonisation
.
Front Immunol
.
2018
;
9
:
2405
.
37.
Voß
F
,
van Beek
LF
,
Schwudke
D
,
Ederveen
THA
,
van Opzeeland
FJ
,
Thalheim
D
, et al
.
Lipidation of pneumococcal antigens leads to improved immunogenicity and protection
.
Vaccines
.
2020
;
8
(
2
):
310
.
38.
Liu
X
,
Kimmey
JM
,
Matarazzo
L
,
de Bakker
V
,
van Maele
L
,
Sirard
J-C
, et al
.
Exploration of bacterial bottlenecks and Streptococcus pneumoniae pathogenesis by CRISPRi-seq
.
Cell Host Microbe
.
2021
;
29
(
1
):
107
20.e6
.
39.
Briles
DE
,
Crain
MJ
,
Gray
BM
,
Forman
C
,
Yother
J
.
Strong association between capsular type and virulence for mice among human isolates of Streptococcus pneumoniae
.
Infect Immun
.
1992
;
60
(
1
):
111
6
.
40.
Ali
MQ
,
Kohler
TP
,
Burchhardt
G
,
Wüst
A
,
Henck
N
,
Bolsmann
R
, et al
.
Extracellular pneumococcal serine proteases affect nasopharyngeal colonization
.
Front Cell Infect Microbiol
.
2020
;
10
:
613467
.
41.
Hermoso
JA
,
Monterroso
B
,
Albert
A
,
Galán
B
,
Ahrazem
O
,
García
P
, et al
.
Structural basis for selective recognition of pneumococcal cell wall by modular endolysin from phage Cp-1
.
Structure
.
2003
;
11
(
10
):
1239
49
.
42.
Wenzel
M
,
Vischer
NOE
,
Strahl
H
,
Hamoen
LW
.
Assessing membrane fluidity and visualizing fluid membrane domains in bacteria using fluorescent membrane dyes
.
Bio Protoc
.
2018
;
8
(
20
):
e3063
.
43.
Wojda
I
,
Staniec
B
,
Sułek
M
,
Kordaczuk
J
.
The greater wax moth Galleria mellonella: biology and use in immune studies
.
Pathog Dis
.
2020
;
78
(
9
):
ftaa057
.
44.
Ménard
G
,
Rouillon
A
,
Cattoir
V
,
Donnio
P-Y
.
Galleria mellonella as a suitable model of bacterial infection: past, present and future
.
Front Cell Infect Microbiol
.
2021
;
11
:
782733
.
45.
Yu
X-Q
,
Kanost
MR
.
Binding of hemolin to bacterial lipopolysaccharide and lipoteichoic acid. An immunoglobulin superfamily member from insects as a pattern-recognition receptor
.
Eur J Biochem
.
2002
;
269
(
7
):
1827
34
.
46.
Junges
R
,
Maienschein-Cline
M
,
Morrison
DA
,
Petersen
FC
.
Complete genome sequence of Streptococcus pneumoniae serotype 19F strain EF3030
.
Microbiol Resour Announc
.
2019
;
8
(
19
):
e00198-19
.
47.
Cuypers
F
,
Schäfer
A
,
Skorka
SB
,
Surabhi
S
,
Tölken
LA
,
Paulikat
AD
, et al
.
Innate immune responses at the asymptomatic stage of influenza A viral infections of Streptococcus pneumoniae colonized and non-colonized mice
.
Sci Rep
.
2021
;
11
(
1
):
20609
.
48.
Briles
EB
,
Tomasz
A
.
Pneumococcal forssman antigen
.
J Biol Chem
.
1973
;
248
(
18
):
6394
7
.
49.
Baumgart
T
,
Hunt
G
,
Farkas
ER
,
Webb
WW
,
Feigenson
GW
.
Fluorescence probe partitioning between Lo/Ld phases in lipid membranes
.
Biochim Biophys Acta
.
2007
;
1768
(
9
):
2182
94
.
50.
Vollmer
W
,
Blanot
D
,
de Pedro
MA
.
Peptidoglycan structure and architecture
.
FEMS Microbiol Rev
.
2008
;
32
(
2
):
149
67
.
51.
Silhavy
TJ
,
Kahne
D
,
Walker
S
.
The bacterial cell envelope
.
Cold Spring Harb Perspect Biol
.
2010
;
2
(
5
):
a000414
.
52.
Rohde
M
.
The Gram-positive bacterial cell wall
.
Microbiol Spectr
.
2019
;
7
(
3
).
53.
Al-Adwani
S
,
Wallin
C
,
Balhuizen
MD
,
Veldhuizen
EJA
,
Coorens
M
,
Landreh
M
, et al
.
Studies on citrullinated LL-37: detection in human airways, antibacterial effects and biophysical properties
.
Sci Rep
.
2020
;
10
(
1
):
2376
.
54.
Sanz
JM
,
Garcia
JL
.
Structural studies of the lysozyme coded by the pneumococcal phage Cp-1. Conformational changes induced by choline
.
Eur J Biochem
.
1990
;
187
(
2
):
409
16
.
55.
Gründling
A
,
Schneewind
O
.
Synthesis of glycerol phosphate lipoteichoic acid in Staphylococcus aureus
.
Proc Natl Acad Sci U S A
.
2007
;
104
(
20
):
8478
83
.
56.
Santa Maria
JP
,
Sadaka
A
,
Moussa
SH
,
Brown
S
,
Zhang
YJ
,
Rubin
EJ
, et al
.
Compound-gene interaction mapping reveals distinct roles for Staphylococcus aureus teichoic acids
.
Proc Natl Acad Sci U S A
.
2014
;
111
(
34
):
12510
5
.
57.
Bharatiya
B
,
Wang
G
,
Rogers
SE
,
Pedersen
JS
,
Mann
S
,
Briscoe
WH
.
Mixed liposomes containing Gram-positive bacteria lipids: lipoteichoic acid (LTA) induced structural changes
.
Colloids Surf B Biointerfaces
.
2021
;
199
:
111551
.
58.
Kandasamy
SK
,
Larson
RG
.
Molecular dynamics simulations of model trans-membrane peptides in lipid bilayers: a systematic investigation of hydrophobic mismatch
.
Biophys J
.
2006
;
90
(
7
):
2326
43
.
59.
Zhang
Y-M
,
Rock
CO
.
Membrane lipid homeostasis in bacteria
.
Nat Rev Microbiol
.
2008
;
6
(
3
):
222
33
.
60.
Tseng
H-J
,
McEwan
AG
,
Paton
JC
,
Jennings
MP
.
Virulence of Streptococcus pneumoniae: PsaA mutants are hypersensitive to oxidative stress
.
Infect Immun
.
2002
;
70
(
3
):
1635
9
.
61.
Lane
JR
,
Tata
M
,
Yasmin
R
,
Im
H
,
Briles
DE
,
Orihuela
CJ
.
PspA-mediated aggregation protects Streptococcus pneumoniae against desiccation on fomites
.
mBio
.
2023
;
14
(
6
):
e0263423
.
62.
Huan
Y
,
Kong
Q
,
Mou
H
,
Yi
H
.
Antimicrobial peptides: classification, design, application and research progress in multiple fields
.
Front Microbiol
.
2020
;
11
:
582779
.
63.
Jenssen
H
,
Hamill
P
,
Hancock
REW
.
Peptide antimicrobial agents
.
Clin Microbiol Rev
.
2006
;
19
(
3
):
491
511
.
64.
Bahar
AA
,
Ren
D
.
Antimicrobial peptides
.
Pharmaceuticals
.
2013
;
6
(
12
):
1543
75
.
65.
Ridyard
KE
,
Overhage
J
.
The potential of human peptide LL-37 as an antimicrobial and anti-biofilm agent
.
Antibiot
.
2021
;
10
(
6
):
650
.
66.
Draing
C
,
Pfitzenmaier
M
,
Zummo
S
,
Mancuso
G
,
Geyer
A
,
Hartung
T
, et al
.
Comparison of lipoteichoic acid from different serotypes of Streptococcus pneumoniae
.
J Biol Chem
.
2006
;
281
(
45
):
33849
59
.
67.
Mücke
P-A
,
Ostrzinski
A
,
Hammerschmidt
S
,
Maaß
S
,
Becher
D
.
Proteomic adaptation of Streptococcus pneumoniae to the antimicrobial peptide human beta defensin 3 (hBD3) in comparison to other cell surface stresses
.
Microorganisms
.
2020
;
8
(
11
):
1697
.
68.
Damjanovic
M
,
Kharat
AS
,
Eberhardt
A
,
Tomasz
A
,
Vollmer
W
.
The essential tacF gene is responsible for the choline-dependent growth phenotype of Streptococcus pneumoniae
.
J Bacteriol
.
2007
;
189
(
19
):
7105
11
.
69.
García
E
,
García
JL
,
García
P
,
Arrarás
A
,
Sánchez-Puelles
JM
,
López
R
.
Molecular evolution of lytic enzymes of Streptococcus pneumoniae and its bacteriophages
.
Proc Natl Acad Sci U S A
.
1988
;
85
(
3
):
914
8
.
70.
Howard
LV
,
Gooder
H
.
Specificity of the autolysin of Streptococcus (Diplococcus) pneumoniae
.
J Bacteriol
.
1974
;
117
(
2
):
796
804
.
71.
De Las Rivas
B
,
García
JL
,
López
R
,
García
P
.
Purification and polar localization of pneumococcal LytB, a putative endo-beta-N-acetylglucosaminidase: the chain-dispersing murein hydrolase
.
J Bacteriol
.
2002
;
184
(
18
):
4988
5000
.
72.
Maestro
B
,
Sanz
JM
.
Choline binding proteins from Streptococcus pneumoniae: a dual role as enzybiotics and targets for the design of new antimicrobials
.
Antibiot
.
2016
;
5
(
2
):
21
.
73.
Pérez-Dorado
I
,
González
A
,
Morales
M
,
Sanles
R
,
Striker
W
,
Vollmer
W
, et al
.
Insights into pneumococcal fratricide from the crystal structures of the modular killing factor LytC
.
Nat Struct Mol Biol
.
2010
;
17
(
5
):
576
81
.
74.
Rennemeier
C
,
Hammerschmidt
S
,
Niemann
S
,
Inamura
S
,
Zähringer
U
,
Kehrel
BE
.
Thrombospondin-1 promotes cellular adherence of Gram-positive pathogens via recognition of peptidoglycan
.
FASEB J
.
2007
;
21
(
12
):
3118
32
.
75.
Schulz
C
,
Gierok
P
,
Petruschka
L
,
Lalk
M
,
Mäder
U
,
Hammerschmidt
S
.
Regulation of the arginine deiminase system by ArgR2 interferes with arginine metabolism and fitness of Streptococcus pneumoniae
.
mBio
.
2014
;
5
(
6
):
e01858-14
.