Open access
Host-Microbial Interactions
Research Article
9 April 2024

Neisseria gonorrhoeae scavenges host sialic acid for Siglec-mediated, complement-independent suppression of neutrophil activation

ABSTRACT

Gonorrhea, caused by the bacterium Neisseria gonorrhoeae (Gc), is characterized by neutrophilic influx to infection sites. Gc has developed mechanisms to resist killing by neutrophils that include modifications to its surface lipooligosaccharide (LOS). One such LOS modification is sialylation: Gc sialylates its terminal LOS sugars with cytidine-5′-monophosphate-N-acetylneuraminic acid, which is scavenged from the host using LOS sialyltransferase (Lst) since Gc cannot make its sialic acid. Sialylation enables sensitive strains of Gc to resist complement-mediated killing in a serum-dependent manner. However, little is known about the contribution of sialylation to complement-independent, direct Gc-neutrophil interactions. In the absence of complement, we found sialylated Gc expressing opacity-associated (Opa) proteins decreased the oxidative burst and granule exocytosis from primary human neutrophils. In addition, sialylated Opa+ Gc survived better than vehicle treated or Δlst Gc when challenged with neutrophils. However, Gc sialylation did not significantly affect Opa-dependent association with or internalization of Gc by neutrophils. Previous studies have implicated sialic acid-binding immunoglobulin-type lectins (Siglecs) in modulating neutrophil interactions with sialylated Gc. Blocking neutrophil Siglecs with antibodies that bind to their extracellular domains eliminated the ability of sialylated Opa+ Gc to suppress the oxidative burst and resist neutrophil killing. These findings highlight a new role for sialylation in Gc evasion of human innate immunity, with implications for the development of vaccines and therapeutics for gonorrhea.

IMPORTANCE

Neisseria gonorrhoeae, the bacterium that causes gonorrhea, is an urgent global health concern due to increasing infection rates, widespread antibiotic resistance, and its ability to thwart protective immune responses. The mechanisms by which Gc subverts protective immune responses remain poorly characterized. One way N. gonorrhoeae evades human immunity is by adding sialic acid that is scavenged from the host onto its lipooligosaccharide, using the sialyltransferase Lst. Here, we found that sialylation enhances N. gonorrhoeae survival from neutrophil assault and inhibits neutrophil activation, independently of the complement system. Our results implicate bacterial binding of sialic acid-binding lectins (Siglecs) on the neutrophil surface, which dampens neutrophil antimicrobial responses. This work identifies a new role for sialylation in protecting N. gonorrhoeae from cellular innate immunity, which can be targeted to enhance the human immune response in gonorrhea.

INTRODUCTION

Neisseria gonorrhoeae (Gc) causes the bacterial sexually transmitted infection gonorrhea. Gc is designated as an urgent threat level pathogen by the Centers for Disease Control and Prevention, with approximately 82.4 million cases each year globally (1). Factors contributing to the prevalence of gonorrhea include increasing resistance to antibiotics, resistance to soluble and cellular innate immune components, and the lack of protective immunity elicited by prior infection (2, 3). Finding new approaches to treat or prevent Gc infection is of utmost importance.
The most abundant surface component on Gc is lipooligosaccharide (LOS). The length and composition of LOS is regulated by lgt (LOS glycosyltransferase) genes. Phase variation of lgt genes results in varying oligosaccharide compositions within a Gc population, including during infection (47). Gc isolated from uncomplicated urethral infection predominantly produces LOS that can be sialylated. Sialylation is the incorporation of sialic acid onto the alpha and/or beta chain terminal galactose of LOS, catalyzed by Gc LOS sialyltransferase (Lst) (810). Gc cannot synthesize sialic acid and instead scavenges cytidine-5′-monophosphate-N-acetylneuraminic acid (CMP-NANA from the host) (11). Lst is constitutively expressed (12, 13), is required for optimal genital tract infection (1416), and uses diverse forms of CMP-sialic acid (17).
LOS sialylation is a form of molecular mimicry that is thought to enable Gc to evade immune recognition (18, 19). Sialylation was originally characterized due to conferring “unstable” serum resistance on Gc recovered from urethral secretions but not after laboratory passage (11, 20, 21). Sialylation has since been shown to inhibit all three complement activation pathways by decreasing C1 engagement and C4b deposition and increasing recruitment of factor H on Gc (17, 22, 23). Sialylated glycans enable the immune system to discriminate self from non-self, achieved in part through sialic acid-binding immunoglobulin-type lectins (Siglecs) (2427). Siglecs are produced by most immune cells, including neutrophils, which are the predominant immune cells recruited in Gc infections and constitute the purulence in gonorrheal secretions (28). Human neutrophils express Siglec-5, -9, and -14. Siglec-5 and -9 contain cytoplasmic immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that recruit SH2 domain-containing phosphatases to inhibit cellular activation (2934). By contrast, Siglec-14 associates with the adapter protein DAP12, which contains an immunoreceptor tyrosine-based activating motif (ITAM) that once phosphorylated, recruits Syk kinase to activate downstream signaling (35, 36). The genes that encode Siglec-5 and Siglec-14 are adjacent and considered paired receptors since the proteins share almost complete sequence identity in their binding domains and similar glycan binding preferences (35, 37). 10%–70% of individuals in certain racial/ethnic groups harbor a SIGLEC14/5 fusion gene where Siglec-14 is not expressed but Siglec-5 is (38). Thus, on balance, signaling through neutrophil Siglecs dampens inflammatory signaling. Sialylated Gc has been reported to interact with the extracellular domains of these Siglecs as Fc chimeras (39).
Gc expresses numerous gene products that defend against neutrophil antimicrobial activities (28). Gc also varies its ability to interact non-opsonically with neutrophils through phase-variable expression of opacity-associated (Opa) proteins (40). Most Opa proteins bind one or more carcinoembryonic antigen-related cell adhesion molecules (CEACAMs). In particular, binding to the granulocyte-restricted CEACAM3 elicits phagocytosis, reactive oxygen species production, and granule release (4147).
In this study, we unexpectedly found that sialylation promotes Gc survival from human neutrophils in a complement-independent manner. Our results implicate Siglecs in dampening the neutrophil activation that is elicited by CEACAM-binding Opa proteins, bringing new insight into the ways that Gc modulates soluble and cellular innate immunity to persist in its obligate human host.

RESULTS

Gc Lst incorporates sialic acid onto LOS, which is retained upon infection of neutrophils

To investigate the role of LOS sialylation on direct, nonopsonic Gc interactions with neutrophils, we first assessed Lst- and CMP-NANA-dependent sialylation of Opa-expressing (Opa+) Gc. The wild-type (WT) Lst parent for these studies was a derivative of strain FA1090 1-81-S2 that constitutively expresses the OpaD protein in an otherwise Opa-negative background, hereafter termed OpaD (48). The OpaD protein binds CEACAMs 1 and 3 on human neutrophils, leading to rapid phagocytic killing (48, 49). This strain produces the sialylatable lacto-N-neotetraose (LNnT) alpha chain and lactose beta chain (50). OpaD was transformed with insertionally inactivated lst to generate an isogenic mutant that cannot be sialylated (OpaDΔlst) (15). Bacteria were incubated with CMP-NANA or vehicle control, then washed to remove residual sialic acid and processed for downstream analysis.
Monoclonal antibody (mAb) 6B4 binds the LNnT epitope of the alpha chain of Neisseria LOS; however, sialylation of its terminal galactose prevents 6B4 binding (8, 51). By Western blot, 6B4 reacted with lysates of OpaD, and binding was lost when OpaD was incubated with CMP-NANA (Fig. 1A). 6B4 reacted with OpaDΔlst lysates with or without CMP-NANA (Fig. 1A). However, the small size (<5 kDa) of LOS makes resolving and detecting it by Western blot a challenge. Therefore, to analyze sialylation on individual bacteria, we used imaging flow cytometry with 6B4. The fluorescence index of OpaD incubated with CMP-NANA was significantly lower than OpaD without CMP-NANA and OpaDΔlst with or without CMP-NANA; these three were statistically indistinguishable (Fig. 1B and C).
Fig 1
The analysis of OpaD with and without the lst gene, with and without addition of sialic acid as CMP-NANA or CMP-Az-NANA, is shown using immunoblot and fluorescence imaging.
Fig 1 Analysis and quantification of Lst-dependent sialylation of gonococcal lipooligosaccharide. (A) OpaD and OpaDΔlst were grown with 50 µg/mL CMP-Neu5Ac (NANA) (+) or vehicle (Veh) (–). Bacterial lysates were Western blotted with mAb 6B4, or anti-Zwf (glucose 6-phosphate dehydrogenase) as loading control, followed by HRP-conjugated secondary antibodies. The blot is representative of n = 3 biological replicates. (B-C) OpaD and OpaDΔlst were incubated with CMP-NANA or vehicle as in A, then stained with Tag-IT Violet (TIV). Bacteria were incubated with 6B4 followed by AlexaFluor647-coupled secondary antibody, fixed, and analyzed by imaging flow cytometry. (B) Average fluorescence index (Fl: mean fluorescence intensity (MFI) x percent positive) from n = 3 biological replicates with 20,000 events collected per condition; each symbol indicates one matched biological replicate. Statistical analysis by one-way ANOVA with Tukey’s multiple comparisons test. **P < .01. (C) One representative image from imaging flow cytometry for each treatment condition and each fluorescence channel is shown. AF647-6B4 signal is a false-colored orange, and yellow numbers indicate the object’s fluorescence intensity value. (D-G) Gc were incubated with either CMP-NANA and/or CMP-Az-NANA at 100 ng/mL. Bacteria were stained with TIV, fixed, and then subjected to copper-catalyzed click chemistry azide-alkyne cycloaddition using fluorescein isothiocyanate (FITC)-alkyne. (D) Representative images of CMP-NANA and CMP-Az-NANA sialylated OpaD, each of which was subjected to alkyne-FITC cycloaddition. Yellow numbers are as in C. (E) OpaD or OpaDΔlst were incubated with indicated concentrations of CMP-Az-NANA or CMP-NANA. The MFI of each condition was quantified from flow cytometry, n = 1. (F) WT OpaD was incubated with 0.1 µg/mL of CMP-Az-NANA along with the indicated concentration of unmodified CMP-NANA. Results are graphed as the MFI of each condition and fit with a non-linear regression line. (G) OpaD sialylated with CMP-Az-NANA was stained with TIV and used to infect IL-8-treated, adherent primary human neutrophils for 30 min. After fixation, extracellular Gc was labeled with anti-PorB and AF647-coupled secondary antibody (false-colored cyan) before permeabilization with 0.1% saponin followed by copper click chemistry. Representative images of infected neutrophils with intracellular (TIV+AF647−) and extracellular (TIV+AF647+) Gc with azide-sialylated LOS that is clicked with the alkyne-FITC probe (FITC+). In C and D, images from the indicated fluorescence channels were exported, false-colored, and cropped for presentation purposes. In G, brightfield and fluorescence channels from each cell were exported from the imaging flow cytometer as a combined image. White lines were added between the images from each channel for clarity.
Beyond LNnT, Gc LOS can be sialylated on its Pk-like alpha chain (18, 52) and the lactose-bearing beta chain (50, 53). To directly detect Lst-dependent sialylation at any site on Gc LOS, independent of 6B4 exclusion, we applied copper click chemistry using azido-labeled sialic acid, followed by alkyne-FITC cycloaddition (5457). OpaD incorporated CMP-Azido (Az)-NANA in a Lst-dependent manner (Fig. 1D and E), which competed with unmodified CMP-NANA at approximately equimolar concentration (Fig. 1F). Using Gc preincubated with CMP-Az-NANA, we monitored the retention of sialylation on OpaD in the presence of neutrophils. Representative images collected at 30 min post-infection showed alkyne-FITC-labeled bacteria that were both intracellularly and extracellularly attached to neutrophils (Fig. 1G). Thus imaging flow cytometry, in combination with immunofluorescence and click chemistry, can be used to track and quantify Gc LOS sialylation alone and in cells.

Lst-dependent sialylation of Opa+ Gc dampens human neutrophil activation and protects Gc from killing by neutrophils

We explored the contribution of Gc Lst-dependent sialylation to neutrophil activation. First, the release of reactive oxygen species (ROS) was measured. As previously reported, non-sialylated OpaD elicited robust neutrophil ROS production (Fig. 2A and B) (42, 45, 48). Sialylation significantly reduced the magnitude of the OpaD-elicited oxidative burst; ROS production was inversely correlated with the concentration of CMP-NANA for sialylation (Fig. 2A and B). Sialylation similarly reduced the oxidative burst in response to other Opa+ bacteria (Fig. 2C; Fig. S1A). OpaDΔlst elicited neutrophil ROS production regardless of CMP-NANA addition and was indistinguishable from non-sialylated OpaD (Fig. 2D; Fig. S1B).
Fig 2
Six graphs depict the reduced reaction of neutrophils to Opa+ bacteria when more sialic acid is added, which requires Lst. Analysis with and without CMP-NANA is done across time.
Fig 2 Sialylation dampens neutrophil activation in response to Opa+ bacteria. (A-B) OpaD incubated with the indicated concentrations of CMP-NANA or vehicle (0 µg/mL) were exposed to primary human neutrophils in the presence of luminol at a multiplicity of infection (MOI) of 100. Neutrophil oxidative burst was measured as relative light units (RLUs) of luminol-dependent chemiluminescence every 3 min over 1 h. (A) displays one representative graph of n = 4; (B) is the average ±SEM area under the curve (AUC) for each condition across replicates (symbols indicate matched biological replicates). (C) Opa60 Gc (blue) or OpaI Gc (orange) were incubated with 50 µg/mL CMP-NANA (+) or vehicle (−) before neutrophil exposure; AUCs of n = 4 biological replicates. Non-stimulatory Opaless Gc (dark gray) exposed neutrophils or neutrophils (PMNs) in luminol alone (light gray) serve as negative controls. (D) WT (purple) or Δlst (red) OpaD were treated with or without CMP-NANA and then added to neutrophils as above; AUCs of n = 3 biological replicates. Statistical analyses by one-way ANOVA with Tukey’s multiple comparisons test. *P < .05; **P < .01. (E-F) OpaD treated with CMP-NANA or vehicle as above was added to adherent, IL-8 primed neutrophils for 15 or 30 min at an MOI of 1. Neutrophils were stained for viability (Zombie Near Infrared) with a PE-coupled antibody against primary granule protein CD63 (E) and an APC-coupled antibody against secondary granule protein CD66b (F) on the cell surface. After fixation, cells were analyzed via spectral flow cytometry. Data are presented as median fluorescence (Med Fl) of PE+ (E) and APC+ (F) live cells, gated using unstained and isotype controls. Results are from n = 4 biological replicates (symbol matched). Statistical analyses were performed by two-way ANOVA with Šídák’s multiple comparisons test. *P < .05.
While ROS does not directly contribute to neutrophil anti-gonococcal activity (5860), ROS production requires intracellular signaling events and granule release (61). Neutrophil granules contain antimicrobial components, some of which are bactericidal for Gc (6264). Thus we analyzed the effect of sialylation on the ability of OpaD to stimulate exocytosis of primary and secondary neutrophil granules as measured by CD63 and CD66b surface presentation, respectively (42). Sialylation of Gc significantly reduced neutrophil exocytosis of primary (Fig. 2E) and secondary (Fig. 2F) granules at 30 min post-infection. Together, these data indicate sialylation reduces the ability of Gc to activate neutrophils.
We next tested the contribution of Lst-dependent sialylation to bacterial survival in the presence of adherent, IL-8-primed primary human neutrophils without the addition of human serum (complement). Sialylated OpaD had a statistically significant increase in survival over non-sialylated bacteria at 15 and 30 min post-infection (Fig. 3A). OpaDΔlst phenocopied non-sialylated OpaD at these times, showing that both sialic acid and Lst were required for the increased survival of OpaD exposed to neutrophils. By 60 and 120 min post-infection, survival of sialylated OpaD was not statistically different from non-sialylated Gc (Fig. 3A). Sialylation also significantly increased the survival from neutrophils of CEACAM-binding Opa60 Gc, through 2-h infection (Fig. 3B). However, survival of Opaless Gc from neutrophils was unaffected by sialylation, which fits with our previous reports that Opa-negative Gc persists in the presence of neutrophils (48, 59, 65) (Fig. 3C). These results highlight a role for Lst-dependent sialylation in survival of Opa+ Gc from neutrophils.
Fig 3
Three graphs depict the percent survival of Opa+ or Opaless bacteria from neutrophils, with and without sialic acid and Lst, over time.
Fig 3 Sialylated, Opa+ bacteria have a survival advantage at early times of neutrophil challenge. Adherent, IL-8 primed neutrophils were infected with sialylated (filled) or non-sialylated (empty) (A) OpaD (purple) or OpaDΔlst (red), at a multiplicity of infection (MOI) of 1. Gc viability was determined by the enumeration of CFUs from lysed neutrophils at the indicated times post-infection, expressed as a percentage of CFU at 0 min. (B-C) Sialylated and non-sialylated Opa60 Gc (blue) (B) and Opaless Gc (gray) (C) were exposed to neutrophils and bacterial viability was measured as in A. Results are presented as the average ± SEM for n > 3 (A) n = 4 (B) or n = 3 (C) biological replicates, matched by symbol within each data set. Statistical comparisons were by mixed effect analysis (A) or two-way ANOVA (B-C) with Holm-Šídák’s multiple comparisons test with the following pairwise significances: *P < .05; **P <  .01; ****P < .0001.

Sialylation of LOS does not directly affect OpaD-CEACAM3 interactions

Given the importance of CEACAM3 to the phagocytic killing of Opa+ Gc in neutrophils, we tested the hypothesis that Gc sialylation disrupted OpaD-CEACAM3 engagement. First, sialylated or non-sialylated OpaD was incubated with a Glutathione S-transferase (GST)-tagged N-terminus of human CEACAM3 (N-CEACAM3), and CEACAM3 binding was measured by flow cytometry using anti-GST fluorescence (66). Lst-dependent sialylation did not significantly affect OpaD binding of N-CEACAM3 (Fig. 4A). Sialylation also did not affect the interaction of OpaD with N-CEACAM1 (Fig. S2). Opaless Gc, which does not bind CEACAMs, served as a negative control. Second, the association of sialylated or non-sialylated OpaD with human CEACAM3-transfected CHO cells was measured by imaging flow cytometry. Lst-dependent sialylation did not significantly affect bacterial association with hCEACAM3-CHO cells (Fig. 4B). These results indicate that contrary to our hypothesis, sialylation does not affect Opa protein-CEACAM engagement.
Fig 4
Four graphs feature OpaD with and without CMP-NANA and the lst gene, and Opaless, with two techniques that indicate no significant difference in binding to CEACAM3.
Fig 4 Sialylation does not affect the binding of OpaD bacteria to CEACAM3. (A) OpaD grown with CMP-NANA or vehicle was incubated with GST-tagged recombinant N-terminal domain of CEACAM3 (N-CEACAM3). The binding of N-CEACAM3 to Gc was detected using a mouse anti-GST antibody, followed by a goat anti-mouse IgG AF488-conjugated antibody. Gc were fixed, stained with DAPI, and analyzed by imaging flow cytometry, to calculate the percent of singlet Gc that are AF488+. Opaless Gc that does not bind CEACAM is shown as a negative control (gray). (B) CHO cells transfected with human CEACAM3 (hCCM3) or empty control vector (Ctrl Vec) were infected with TIV-labeled OpaD, and treated with CMP-NANA or vehicle. After 30 min, cells were collected, stained with a pan-CEACAM antibody followed by an AF555-conjugated secondary antibody, and fixed. The percent of singlet CHO cells that are TIV+ (infected) was calculated using imaging flow cytometry. Results are from n = 3 biological replicates (symbol matched). Statistical comparisons were by two-way ANOVA with Tukey’s multiple comparisons test; not significant. (C-D) OpaD with or without sialylated LOS were labeled with TIV and used to infect adherent, IL-8 primed neutrophils. At the indicated time points, infected cells were fixed and extracellular Gc was detected with an anti-PorB antibody, followed by an AF488-coupled secondary antibody, without permeabilization. Cells were then analyzed using imaging flow cytometry to report the percent of neutrophils with associated (%TIV+) Gc (C) or internalized (%TIV+ AF488-) Gc (D). Results are from n = 6 biological replicates (symbol matched). Statistical comparisons were by two-way ANOVA with Tukey’s multiple comparisons test; not significant.
In a previous report, sialylation decreased the adherence of Opa+ Gc to neutrophils in suspension, but this effect was lost over time (67). We thus hypothesized that the mechanism by which sialylated Gc resisted killing by neutrophils was due to decreased association with and/or internalization by these immune cells. To test this hypothesis, we used imaging flow cytometry (68), in which extracellular Gc was discriminated from intracellular bacteria by the accessibility of a mAb against FA1090 PorB1b. The binding of this antibody to Gc was unaffected by sialylation (Fig. S3). Sialylation did not significantly affect OpaD association with (Fig. 4C) or internalization by (Fig. 4D) neutrophils through 30 min post-infection, when effects were measured by ROS production and bacterial survival. We conclude that sialylation does not directly affect Opa+ Gc interaction with neutrophil CEACAMs.

Sialylated Gc binds immunoregulatory Siglecs to dampen neutrophil activation and anti-gonococcal activity

The inhibitory effect of Gc sialylation on neutrophil activation and not phagocytosis, along with work from other groups (37, 39, 69, 70), led us to investigate the contribution of sialic acid immunoglobulin-type lectins (Siglecs) on neutrophils. Using imaging flow cytometry, we confirmed the presence of Siglec-9 and Siglec-5/14 on the surface of primary human neutrophils (Fig. 5A) compared to unstained (Fig. S4A). Staining ranged from small puncta to rings around the cell periphery, though there was minimal overlap in Siglec-9 vs Siglec-5/14 localization (Fig. 5B).
Fig 5
Graphs and micrographs show that antibodies against Siglec 9 and Siglec 5/14 increase neutrophil activation and decrease Opa+ Gc survival.
Fig 5 Blockade of neutrophil Siglec-9 and Siglec-5/14 reverses the ability of sialylated gonococci to suppress neutrophil activation and antibacterial activity. (A-B) Uninfected and adherent, IL-8 primed human neutrophils were incubated with antibodies that recognize human Siglec-9-APC and/or Siglec-5/Siglec-14-AF488, without cell permeabilization before fixation and processing via imaging flow cytometry. (A) Dot plot of focused singlet neutrophils with anti-Siglec-9(APC) and anti-Siglec-5/14(AF488). The X-axis indicates AF488 fluorescence intensity and the y-axis indicates APC fluorescence intensity. Double positive (green) gate based on single stains; number within the gate is the percent of the stained condition. (B) Representative images of neutrophils stained with anti-Siglec-9-APC (cyan) and anti-Siglec-5/14-AF488 (yellow); colored fluorescence in merged. Brightfield and fluorescence channels from each cell were exported from the imaging flow cytometer as a combined image. White lines were added between the images from each channel for clarity. Results are representative of n = 3 biological replicates by spectral flow cytometry and confocal microscopy (not shown) and demonstrate n = 1 replicate with imaging flow cytometry. (C-D) Primary human neutrophils were incubated with anti-human Siglec-9 and Siglec-5/-14 antibodies (green) or media alone (purple) for 30 min. (C) Neutrophils were exposed to sialylated (filled bars) or non-sialylated (open bars) OpaD at an MOI of 100 in the presence of luminol. ROS production was measured over 1 h by luminol-dependent chemiluminescence and presented as an area under the curve for each condition across n = 6 biological replicates (symbol matched). Statistical comparisons by one-way ANOVA with Tukey’s multiple comparisons test. *P < .05. (D) Neutrophils were treated with anti-human Siglec-9 and Siglec-5/-14 antibodies (green) or media alone (purple) as in C. Cells were then infected with sialylated (filled) or non-sialylated (empty) Gc at an MOI of 1. Survival of Gc is presented as a percent of the bacteria at time 0 min, as in Fig. 3. Statistical analyses by two-way ANOVA with Holm-Šídák’s multiple comparisons test. n = 4 biological replicates (symbol matched); *P ≤ .05, ***P < 0.0001.
We hypothesized that sialic acid-Siglec engagement interfered with Opa-CEACAM binding to dampen the neutrophil response to Opa+ Gc. To test this, neutrophils were incubated with blocking antibodies against the extracellular domains of Siglec-9 and -5/14 (71), then exposed to sialylated or non-sialylated OpaD, and ROS production was measured. The addition of anti-Siglec antibodies significantly enhanced the neutrophil oxidative burst elicited by sialylated OpaD (solid green vs solid purple, Fig. 5C; Fig. S4B), restoring it to levels that were no different from unsialylated OpaD (open purple bar, Fig. 5C). The effect of Siglec-blocking antibodies on ROS production was specific to sialylated Gc since non-sialylated OpaD elicited similar levels of ROS from neutrophils whether or not the antibodies were present (green outline vs purple outline, Fig. 5C). These data implicate Siglecs in the ability of sialylated Opa+ Gc to reduce neutrophil activation.
The dampened ROS release in response to sialylated Opa+ Gc led us to examine whether blocking Siglecs restored the ability of neutrophils to control Opa+ bacteria. Neutrophils were incubated with blocking antibodies against Siglec-9 and -5/14 or left untreated, then exposed to sialylated or non-sialylated OpaD. The addition of anti-Siglec antibodies significantly reduced the survival of sialylated OpaD after infection of neutrophils at 15 and 30 min, to a level equivalent to non-sialylated OpaD (Fig. 5D). Non-sialylated OpaD were recovered similarly from neutrophils whether or not anti-Siglec antibodies were present, indicating that the effect of Siglecs on Gc-neutrophil interaction is specific for sialylated Gc.
From these results, we conclude that Gc uses sialylation of its LOS in a Siglec-mediated, complement-independent manner to impede neutrophil activation and anti-gonococcal responses, thereby promoting bacterial survival during infection.

DISCUSSION

Gc sialylation of LOS by Lst is crucial for complement resistance and pathogenesis in vivo (10). This study reveals an unexpected, complement-independent role of sialylation: its ability to restrain neutrophil activation in response to Opa+ Gc, improving Gc survival upon neutrophil challenge. Sialylation reduced granule mobilization and release of both oxidative and nonoxidative species in response to Opa+ bacteria. Unexpectedly, sialylation of the LOS did not have a significant effect on Opa-CEACAM binding and engagement. Instead, by blocking the immunoregulatory Siglecs on neutrophils, sialylated Opa+ Gc could no longer suppress neutrophil activation or killing capacity. These data suggest the exploitation of neutrophil self-associated molecular pattern recognition by Gc through LOS sialylation by Lst. Our findings add to the increasing understanding of how pathogens co-opt host factors to impair immune cell activation (72).
This study focused on Gc interactions with neutrophils, the main cell type found in human infectious secretions. We used human neutrophils that were adherent and treated with IL-8, mimicking post-migration behavior in these terminally differentiated, short-lived primary cells (59). Work by our group and others has investigated how Gc expression of phase-variable Opa proteins affects their survival from neutrophils (4042, 45, 48, 63, 73). Most Opa proteins bind selected human CEACAMs. In particular, neutrophils and other granulocytes express CEACAM3, which contains an ITAM that can recruit the Src family and Syk tyrosine kinases to drive signaling pathways leading to phagocytosis, granule release, and ROS production (40). Given the negative charge conferred by sialic acids, we hypothesized that LOS sialylation would impair the phagocytic activity of neutrophils toward Gc. However, we found no significant difference in the ability of sialylated Opa+ Gc to bind the soluble N-domain of CEACAM, CEACAM-expressing cells, or primary human neutrophils. While our study only focused on Opa+ Gc that bind CEACAMs 1 and 3, future work can explore how sialylation affects other modes of interaction between Gc and neutrophils, including antibody- and complement-mediated association.
Siglecs help the innate immune system to distinguish between non-self and self-associated molecular patterns (26). Many immune cell Siglecs contain cytoplasmic ITIM domains that transduce inhibitory signals to dampen inflammation (24). Other sialylated bacteria, including N. meningitidis and Group B Streptococcus, have been shown to manipulate cell activation via Siglec engagement (27, 69), and Gc can bind human Siglec-Fc chimera proteins (39). Consistent with published findings, we detected Siglec-9 and Siglec-5/-14 on the human neutrophil surface (29, 32, 37). Moreover, blocking Siglecs reversed the inhibitory activity of sialylation on neutrophil responses to Gc, uncovering a new role for Siglecs in thwarting neutrophil anti-gonococcal responses. Interestingly, human Siglec-Fc chimeras have also been reported to bind PorB on the surface of unsialylated Gc, which was enhanced by the production of a shorter, nonsialylatable LOS (39). Whether non-sialylated Gc interacts with Siglecs on neutrophils and the consequences of such interaction remain to be explored. Unlike Siglec-5 and Siglec-9, Siglec-14 does not have a cytoplasmic domain; instead, it associates with the ITAM-bearing DAP12 (35, 37). Siglec-5 and Siglec-14 have almost identical ligand binding domains due to ongoing gene conversion and are considered paired receptors, possibly to balance immune responses to pathogens (24, 35, 38). In a study of genetic variations in Siglecs among a human cohort with a high burden of gonorrhea, uninfected individuals were more likely to produce Siglec-14, though not reaching statistical significance (39). Future work can examine whether neutrophils from individuals with or without Siglec-14 respond differently to sialylated Gc.
The localization of Lst has recently been updated from an outer membrane protein to the inner face of the cytoplasmic membrane, where the OS is assembled (74, 75). Unlike the closely related microbe N. meningitidis, Gc cannot endogenously synthesize CMP-NANA and must scavenge it from the extracellular environment for sialylation. Lst has both α2,3 (alpha chain LNnT) and α2,6 sialyltransferase activity (alpha chain Pk-like OS and beta chain lactose) and has been shown to use different CMP-sialic acids as substrates (8, 17, 52, 53). This promiscuity suggests that sialic acid availability rather than enzymatic activity drives the dynamics of LOS sialylation. We exploited this feature of Lst to add azido-labeled CMP-NANA in the form of N-acetylneuraminic acid (Neu5Ac) to directly track sialylation on the Gc surface. Other Lst substrates include CMP-Neu5Gc, which is missing in humans due to the evolutionary loss of the responsible hydrolase (76), and CMP-legionaminic acid and CMP-ketodeoxynonulosonic acid, which do not confer complement resistance and could serve as anti-gonococcal therapeutics (17).
One key question prompted by this work is how neutrophils affect the dynamics of Gc sialylation. Neutrophils have been proposed as a source of CMP-NANA since lysates from polymorphonuclear phagocytes, including neutrophils, confer “unstable” serum resistance on Gc (77). How Gc obtains CMP-NANA, which is cytoplasmic, is currently unknown since intracellular Gc resides in phagolysosomes (65). Phagosomes may contain sialic acid transporters; in addition, neutrophils may release CMP-NANA by lysis or elaboration of neutrophil extracellular traps. It is also tantalizing to speculate that neutrophil sialyltransferases, found in the secretory pathway, could intersect with Gc- and sialic acid-containing compartments to sialylate the bacteria (78). Conversely, neutrophils have surface-exposed sialidases, which desialylate their glycans during diapedesis (79, 80) and other cells’ glycans for adhesion (81). While we did not detect any desialylation of Gc over the first 30 min of neutrophil exposure, the sialylation state of Gc over time with neutrophils remains unknown. Modification of neutrophil-like cells to synthesize azido-labeled CMP-NANA (82) would allow sialylation of Gc to be tracked over time and in different compartments in an Lst-dependent manner.
Sialylated Gc has reduced infectivity in the experimental challenge of the human male urethra (14) and the genital tract of female mice (15, 16). However, Gc isolated from male urethral gonorrheal exudates are sialylated (8, 11, 21). In addition, Gc isolated from cervicovaginal human secretions are less likely to be sialylated, due to sialidase activity conferred by members of the female genital microbiome (83). Given these dynamics, we anticipate that balancing the sialylation state allows Gc to resist cellular and humoral immunity—including by engaging Siglecs to thwart neutrophil attack—while maintaining infectivity of target mucosal surfaces. These findings can be exploited for novel therapies for gonorrhea since Lst expression is not a phase variable. For instance, CMP-NANA analogs that do not confer serum resistance aid complement killing of the multidrug-resistant Gc (17, 84). Similarly, sialic acid analogs that do not engage Siglecs could be developed to render Gc more sensitive to neutrophils. These approaches, in conjunction with antibiotics and vaccines, could enhance neutrophil and soluble host defenses to combat drug-resistant gonorrhea.

MATERIALS AND METHODS

Bacterial strains and growth conditions

Gc in this study is in the FA1090 background, constitutively expressing the pilin variant 1-81-S2 and with in-frame deletions of all opa genes (Opaless) (48). Opaless Gc with non-phase variable, constitutively expressed opaD has been described previously (OpaD) (48); Opa60 and OpaI were constructed similarly (49, 66).
Gc was grown overnight on gonococcal medium base (Difco) with Kellogg’s supplements (85) (GCB) at 37°C with 5% CO2. Growth of Gc in a rich liquid medium containing Kellogg’s supplements and NaHCO3 to enrich for piliated, live, mid-logarithmic phase bacteria has been previously described (86). For sialylation, 50 µg/mL of CMP-NANA (Nacalai) reconstituted in phosphate-buffered saline (PBS) was added for the final 2.5-h growth (87). Gc was washed with PBS + 5 mM MgSO4 prior to infection. CMP-Az-NANA (R&D Systems) was added in lieu of or in competition with unmodified CMP-NANA (54).
Isogenic OpaDΔlst was generated by spot-transforming OpaD with genomic DNA from MS11 GP300 lst::Kan (from A. Jerse, USUHS) (15). Transformants were selected on GCB with 50 µg/mL kanamycin and backcrossed three times. Loss of Lst was confirmed by PCR, and the inability to sialylate itself was verified by 6B4 exclusion (see below). OpaD expression was confirmed by Western blotting bacterial lysates with 4B12 mAb against Opa (88, 89).

Gc sialylation and detecting sialylated Gc

Western blot: SDS-PAGE and immunoblotting for 6B4 reactivity were conducted as previously described (17). In brief, lysates from Gc with or without CMP-NANA pre-incubation were resolved using a 4%–20% gradient SDS-polyacrylamide gel (Bio-Rad) and transferred in CAPS methanol buffer to polyvinylidene difluoride (PVDF). Membranes were blocked in PBS + 0.5% Tween-20 +5% dry skim milk and probed with 6B4 tissue culture supernatant (from S. Ram, UMass) followed by anti-mouse IgM-horseradish peroxidase (HRP) antibody (Jackson ImmunoResearch). Blots were developed with SuperSignal chemiluminescent substrate (ThermoFisher). For imaging flow cytometry, Gc (108 CFU/mL), incubated with or without 50 µg/mL CMP-NANA, was labeled with 5 µM Tag-IT Violet (TIV) (BioLegend) and then incubated with 6B4, followed by AlexaFluor647-coupled goat anti-mouse IgM (Jackson ImmunoResearch) before fixation and processing. Samples were examined using ImageStream X Mark II imaging flow cytometer and analyzed using INSPIRE and IDEAS v. 6.2 software packages (Amnis Luminex Corporation). Focused, singlet TIV+ Gc was gated as in reference (66). AF647+ gate was set in reference to TIV+ Gc without 6B4. Data are reported as fluorescence index (geometric mean fluorescence intensity × percent AF647+).
Click chemistry: Gc (108 CFU/mL) with or without CMP-Az-NANA were stained with TIV before fixation to analyze Gc alone, or used to infect primary human neutrophils at a multiplicity of infection (MOI) of 1 (see below). After fixation with 2% paraformaldehyde (PFA), neutrophils were incubated with H.5 mouse anti-PorB (from M. Hobbs, UNC) to detect extracellular Gc, then permeabilized with 0.2% saponin. Copper-catalyzed click chemistry was carried out for 1 h at room temperature with continuous shaking, using reaction reagents: 2.5 mM THPTA (tris-hydroxypropyltriazolylmethylamine) ligand (Lumiprobe), 0.06 mM FAM alkyne 5-isomer (5-Carboxyfluorescein) (Lumiprobe), 2.4 mM L-ascorbic acid (VWR), and 2 mM copper (II) sulfate pentahydrate all dissolved in UltraPure distilled water (Invitrogen) (90). Samples were thoroughly washed before analysis by imaging flow cytometry.

Neutrophil isolation

Human subjects research was conducted in accordance with a protocol approved by the University of Virginia Institutional Review Board for Health Sciences Research (#13909). Informed, formal written consent was obtained from each participant. Neutrophils were isolated from venous blood collected from healthy human subjects via dextran sedimentation followed by a Ficoll gradient as described (86). Neutrophils were resuspended in Dulbecco’s PBS (DPBS; without calcium and magnesium; Thermo Scientific) with 0.1% dextrose and used within 1 h after isolation. Samples were >95% neutrophils and >99% viable.

Neutrophil ROS production

Neutrophils were resuspended in Morse’s Defined Medium (MDM) (91) containing 20 µM luminol. Gc was added at an MOI of 100 and the generation of reactive oxygen species was measured every 3 min for 1 h via chemiluminescence (86). Negative controls included Opaless-infected and uninfected neutrophils (48). Line graphs are representative of three or more biological replicates. Bar graphs present all biological replicates’ area under the curve (AUC) values.

Neutrophil granule exocytosis

Surface expression of neutrophil granule proteins was measured as in reference (42), using phycoerythrin (PE)-CD63 for primary granules and allophycocyanin (APC)-CD66b for secondary granules (or isotype controls PE-IgG1 and APC-IgM) (BioLegend). Data were acquired using a Cytek Northern Lights spectral flow cytometer and analyzed using FCS Express (De Novo Software). The median fluorescence of each sample was normalized to unstimulated neutrophils as negative control.

Gc survival in the presence of neutrophils

Infection of adherent, IL-8 treated human neutrophils in RPMI 1640 medium with 10% heat-inactivated fetal bovine serum (HI-FBS) was conducted as in reference (86), except Gc was pre-incubated with or without CMP-NANA. After centrifugation to synchronize infection and incubation at 37˚C, 5% CO2, neutrophils were lysed with 1% saponin, serially diluted, and plated for overnight growth on GCB. Results are reported as a percentage of CFUs enumerated from lysates at the indicated timepoint, divided by CFUs enumerated at 0 min.

Imaging flow cytometric analysis of Gc association with and internalization by neutrophils

Infection of primary human neutrophils with TIV+ Gc and evaluation by imaging flow cytometry was performed as described (42, 68, 92). Because sialylation reduced bacterial binding of the goat anti-Gc antibody (Biosource) to almost undetectable levels (data not shown), extracellular Gc was recognized instead with H.5 anti-PorB, followed by AF647-coupled goat anti-mouse antibody.

N-CEACAM1 and N-CEACAM3 binding of Opa-expressing Gc

GST-tagged N-terminal domains of human CEACAMs 1 and 3 were purified and utilized as in reference (66). In brief, non-sialylated or sialylated Gc were incubated with GST-N-CEACAM1 or -N-CEACAM3 for 30 min at 37°C with end-over-end rotation. Gc was washed and incubated with mouse anti-GST antibody p1A12 (BioLegend) followed by an AF488-conjugated goat anti-mouse antibody (Jackson ImmunoResearch). The percentage of AF488+ (N-CEACAM-bound) Gc was measured using imaging flow cytometry.

Gc association with CHO-hCEACAM-1 and -3 cells

hCEACAM1-, hCEACAM3-, and Control-CHOs were generated using heparan-sulfate proteoglycan-deficient Chinese hamster ovary (CHO) cell strain PgsD-677 (ATCC). Cells were grown in Ham’s F-12K medium (ATCC) with HI-FBS and 1× Antibiotic-Antimycotic (Gibco) at 37°C, 5% CO2. Plasmids were prepared from VectorBuilder Escherichia coli stocks using Plasmid Miniprep (Qiagen) and ampicillin selection: human CEACAM1 (VB900000-5687rxc), human CEACAM3 (VB900000-7471zkb), or an empty control (VB010000-9288rhy) vector under a CAG promoter. CHOs were transfected using Lipofectamine 3000 transfection reagent (Invitrogen) according to the manufacturer’s instructions. After 48 h, transfectants were selected using 10 µg/mL puromycin (Gibco). A non-transfected control was used to confirm selection. Transfectants were maintained under puromycin selection throughout culturing and passaged at 70%–90% confluency; the medium was replaced with an antibiotic-free medium 2 h prior to infection. Surface expression of CEACAM on pooled CEACAM1- and 3-CHO transfectants and absence on Control-CHOs were confirmed by immunofluorescence microscopy using an anti-Pan-CEACAM antibody (Abcam, clone D14HD11).
Cells were seeded in a six-well plate and grown for 48 h to confluency (~106 cells/well). TIV +Gc were added at an MOI of 1 to cells on ice, centrifuged to synchronize infection, washed, and incubated for 30 min. Cells were lifted with Versene solution (Gibco) at 37°C, 5% CO2 for 10 min. Cells were then fixed with 2% PFA, collected with a rubber cell scraper, and filtered through 50 µm nylon mesh. Cells were blocked in PBS + 10% normal goat serum and incubated with anti-Pan-CEACAM antibody followed by goat anti-mouse AF555-conjugated antibody (Invitrogen). Cells were analyzed using imaging flow cytometry and IDEAS software. Focused cells were gated by gradient root mean square for image sharpness >50. Single cells were gated by a high aspect ratio and area >200 and <1,000. Single stained controls were used to set gates for AF555+ and TIV+ populations.

Siglec staining and blockade

Adherent, IL-8 primed neutrophils were fixed and lifted with a cell scraper then cells were washed in PBS and blocked in 10% normal goat serum. Next, neutrophils were incubated with anti-Siglec-9-APC (Clone 191240) and/or anti-Siglec-5/14-AF488 (Clone 194128) (R&D Systems) for 30 min. Samples were analyzed using imaging flow cytometry as above. To block Siglecs, isolated neutrophils were incubated with the unconjugated forms of anti-Siglec-9 and/or anti-Siglec-5/-14 for 30 min at 37°C + 5% CO2 before infection.

Statistics

Results are depicted as the means ± standard errors for >3 independent experiments. Statistics were calculated using GraphPad Prism; a P value of ≤ 0.05 was considered significant. Analysis of variance (ANOVA) or mixed model analysis was used for multiple comparisons of parametric data.

ACKNOWLEDGMENTS

We thank Louise Ball and Samuel Clark for preliminary experiments that informed this project, and Asya Smirnov and Linda Columbus for guidance in methods development. We thank Mike Solga of the UVA Flow Cytometry Core Facility (RRid: SCR_017829) and Hazel Ozuna (University of Louisville) for advice, Ann Jerse and Marcia Hobbs for strains and antibodies, and Sanjay Ram for reagents and discussions. We are grateful to the human subjects who made this research possible.
This work was supported by NIH R01AI097312, NIH U19AI144180, and NIH U01AI162457 (A.K.C.). A.J.C. was supported in part by NIH F31AI157528 and NIH 3R01AI097312-07S1. A.J.C. and M.W.B. were supported in part by NIH T32AI007046. M.M.P. and N.J.F. were supported by NIH R35GM124893. G.A.J. and C.M.J. were supported by the Merit Review Award BX000727 and the Research Career Scientist Award from the Research Service of the U.S. Department of Veterans Affairs.
Conceptualization: A.J.C. and A.K.C. Formal analysis: A.J.C, K.S.T., and N.J.F. Funding acquisition: A.K.C. and A.J.C. Project Administrator: A. K.C. Investigation: A.J.C., K.S.T., M.W.B., and N.J.F. Methodology: A.J.C., M.W.B., N.J.F., and M.M.P. Supervision: G.A.J., C.M.J., M.M.P., and A.K.C. Visualization: A.J.C. Writing—original draft: A.J.C. and A.K.C. Writing—review & editing: A.J.C., K.S.T., M.W.B., N.J.F., C.M.J., G.A.J., and A.K.C.

SUPPLEMENTAL MATERIAL

Supplemental Figures - mbio.00119-24-s0001.pdf
Figures S1 to S4.
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
World Health Organization. 2024 Gonorrhoea (Neisseria gonorrhoeae infection). Available from: https://www.who.int/news-room/fact-sheets/detail/gonorrhoea-(neisseria-gonorrhoeae-infection)
2.
Workowski KA, Bachmann LH, Chan PA, Johnston CM, Muzny CA, Park I, Reno H, Zenilman JM, Bolan GA. 2021. Sexually transmitted infections treatment guidelines, 2021. MMWR Recomm Rep 70:1–187.
3.
Rice PA, Shafer WM, Ram S, Jerse AE. 2017. Neisseria gonorrhoeae: drug resistance, mouse models, and vaccine development. Annu Rev Microbiol 71:665–686.
4.
Yamasaki R, Bacon BE, Nasholds W, Schneider H, Griffiss JM. 1991. Structural determination of oligosaccharides derived from lipooligosaccharide of Neisseria gonorrhoeae F62 by chemical, enzymatic, and two-dimensional NMR methods. Biochemistry 30:10566–10575.
5.
John CM, Griffiss JM, Apicella MA, Mandrell RE, Gibson BW. 1991. The structural basis for pyocin resistance in Neisseria gonorrhoeae lipooligosaccharides. J Biol Chem 266:19303–19311.
6.
Gotschlich EC. 1994. Genetic locus for the biosynthesis of the variable portion of Neisseria gonorrhoeae lipooligosaccharide. J Exp Med 180:2181–2190.
7.
Tong Y, Arking D, Ye S, Reinhold B, Reinhold V, Stein DC. 2002. Neisseria gonorrhoeae strain PID2 simultaneously expresses six chemically related lipooligosaccharide structures. Glycobiology 12:523–533.
8.
Apicella MA, Mandrell RE, Shero M, Wilson ME, Griffiss JM, Brooks GF, Lammel C, Breen JF, Rice PA. 1990. Modification by sialic acid of Neisseria gonorrhoeae lipooligosaccharide epitope expression in human urethral exudates: an immunoelectron microscopic analysis. J Infect Dis 162:506–512.
9.
Mandrell RE, Smith H, Jarvis GA, Griffiss JM, Cole JA. 1993. Detection and some properties of the sialyltransferase implicated in the sialylation of lipopolysaccharide of Neisseria gonorrhoeae. Microb Pathog 14:307–313.
10.
Ram S, Shaughnessy J, de Oliveira RB, Lewis LA, Gulati S, Rice PA. 2017. Gonococcal lipooligosaccharide sialylation: virulence factor and target for novel immunotherapeutics. Pathog Dis 75:ftx049.
11.
Nairn CA, Cole JA, Patel PV, Parsons NJ, Fox JE, Smith H. 1988. Cytidine 5'-monophospho-N-acetylneuraminic acid or a related compound is the low Mr factor from human red blood cells which induces gonococcal resistance to killing by human serum. J Gen Microbiol 134:3295–3306.
12.
Gilbert M, Watson DC, Cunningham AM, Jennings MP, Young NM, Wakarchuk WW. 1996. Cloning of the lipooligosaccharide alpha-2,3-sialyltransferase from the bacterial pathogens Neisseria meningitidis and Neisseria gonorrhoeae. J Biol Chem 271:28271–28276.
13.
Packiam M, Shell DM, Liu SV, Liu YB, McGee DJ, Srivastava R, Seal S, Rest RF. 2006. Differential expression and transcriptional analysis of the alpha-2,3-sialyltransferase gene in pathogenic Neisseria spp. Infect Immun 74:2637–2650.
14.
Schneider H, Schmidt KA, Skillman DR, Van De Verg L, Warren RL, Wylie HJ, Sadoff JC, Deal CD, Cross AS. 1996. Sialylation lessens the infectivity of Neisseria gonorrhoeae Ms11mkC. J Infect Dis 173:1422–1427.
15.
Wu H, Jerse AE. 2006. Alpha-2,3-sialyltransferase enhances Neisseria gonorrhoeae survival during experimental murine genital tract infection. Infect Immun 74:4094–4103.
16.
Lewis LA, Gulati S, Burrowes E, Zheng B, Ram S, Rice PA. 2015. Alpha-2,3-sialyltransferase expression level impacts the kinetics of lipooligosaccharide sialylation, complement resistance, and the ability of Neisseria gonorrhoeae to colonize the murine genital tract. mBio 6:e02465-14.
17.
Gulati S, Schoenhofen IC, Whitfield DM, Cox AD, Li J, St Michael F, Vinogradov EV, Stupak J, Zheng B, Ohnishi M, Unemo M, Lewis LA, Taylor RE, Landig CS, Diaz S, Reed GW, Varki A, Rice PA, Ram S. 2015. Utilizing CMP-sialic acid analogs to unravel Neisseria gonorrhoeae lipooligosaccharide-mediated complement resistance and design novel therapeutics. PLoS Pathog 11:e1005290.
18.
Mandrell RE, Griffiss JM, Macher BA. 1988. Lipooligosaccharides (LOS) of Neisseria gonorrhoeae and Neisseria meningitidis have components that are Immunochemically similar to precursors of human blood group antigens. carbohydrate sequence specificity of the mouse monoclonal antibodies that recognize crossreacting antigens on LOS and human erythrocytes. J Exp Med 168:107–126.
19.
Mandrell RE. 1992. Further antigenic similarities of Neisseria gonorrhoeae lipooligosaccharides and human glycosphingolipids. Infect Immun 60:3017–3020.
20.
Ward ME, Watt PJ, Glynn AA. 1970. Gonococci in urethral exudates possess a virulence factor lost on subculture. Nature 227:382–384.
21.
Martin PM, Patel PV, Parsons NJ, Smith H. 1982. Induction in gonococci of phenotypic resistance to killing by human serum by human genital secretions. Br J Vener Dis 58:363–365.
22.
Ram S, Sharma AK, Simpson SD, Gulati S, McQuillen DP, Pangburn MK, Rice PA. 1998. A novel sialic acid binding site on factor H mediates serum resistance of sialylated Neisseria gonorrhoeae. J Exp Med 187:743–752.
23.
Gulati S, Sastry K, Jensenius JC, Rice PA, Ram S. 2002. Regulation of the mannan-binding lectin pathway of complement on Neisseria gonorrhoeae by C1-inhibitor and alpha 2-macroglobulin. J Immunol 168:4078–4086.
24.
Pillai S, Netravali IA, Cariappa A, Mattoo H. 2012. Siglecs and immune regulation. Annu Rev Immunol 30:357–392.
25.
Macauley MS, Crocker PR, Paulson JC. 2014. Siglec-mediated regulation of immune cell function in disease. Nat Rev Immunol 14:653–666.
26.
Läubli H, Varki A. 2020. Sialic acid-binding immunoglobulin-like Lectins (Siglecs) detect self-associated molecular patterns to regulate immune responses. Cell Mol Life Sci 77:593–605.
27.
Carlin AF, Chang YC, Areschoug T, Lindahl G, Hurtado-Ziola N, King CC, Varki A, Nizet V. 2009. Group B Streptococcus suppression of phagocyte functions by protein-mediated engagement of human Siglec-5. J Exp Med 206:1691–1699.
28.
Palmer A, Criss AK. 2018. Gonococcal defenses against antimicrobial activities of neutrophils. Trends Microbiol 26:1022–1034.
29.
Cornish AL, Freeman S, Forbes G, Ni J, Zhang M, Cepeda M, Gentz R, Augustus M, Carter KC, Crocker PR. 1998. Characterization of Siglec-5, a novel glycoprotein 529 expressed on myeloid cells related to Cd33. Blood 92:2123–2132.
30.
Avril T, Freeman SD, Attrill H, Clarke RG, Crocker PR. 2005. Siglec-5 (CD170) can mediate inhibitory signaling in the absence of immunoreceptor tyrosine-based inhibitory motif phosphorylation. J Biol Chem 280:19843–19851.
31.
Erickson-Miller CL, Freeman SD, Hopson CB, D’Alessio KJ, Fischer EI, Kikly KK, Abrahamson JA, Holmes SD, King AG. 2003. Characterization of Siglec-5 (Cd170) expression and functional activity of anti-Siglec-5 antibodies on human phagocytes. Exp Hematol 31:382–388.
32.
Avril T, Floyd H, Lopez F, Vivier E, Crocker PR. 2004. The membrane-proximal immunoreceptor tyrosine-based inhibitory motif is critical for the inhibitory signaling mediated by Siglecs-7 and -9, CD33-related Siglecs expressed on human monocytes and NK cells. J Immunol 173:6841–6849.
33.
Lizcano A, Secundino I, Döhrmann S, Corriden R, Rohena C, Diaz S, Ghosh P, Deng L, Nizet V, Varki A. 2017. Erythrocyte sialoglycoproteins engage Siglec-9 on neutrophils to suppress activation. Blood 129:3100–3110.
34.
Delaveris CS, Wilk AJ, Riley NM, Stark JC, Yang SS, Rogers AJ, Ranganath T, Nadeau KC, Stanford C-B, Blish CA, Bertozzi CR. 2021. Synthetic Siglec-9 agonists inhibit neutrophil activation associated with COVID-19. ACS Cent Sci 7:650–657.
35.
Angata T, Hayakawa T, Yamanaka M, Varki A, Nakamura M. 2006. Discovery of Siglec-14, a novel sialic acid receptor undergoing concerted evolution with Siglec-5 in primates. FASEB J 20:1964–1973.
36.
Lanier LL, Bakker ABH. 2000. The ITAM-bearing transmembrane adaptor Dap12 in lymphoid and myeloid cell function. Immunol Today 21:611–614.
37.
Ali SR, Fong JJ, Carlin AF, Busch TD, Linden R, Angata T, Areschoug T, Parast M, Varki N, Murray J, Nizet V, Varki A. 2014. Siglec-5 and Siglec-14 are polymorphic paired receptors that modulate neutrophil and amnion signaling responses to group B Streptococcus. J Exp Med 211:1231–1242.
38.
Yamanaka M, Kato Y, Angata T, Narimatsu H. 2009. Deletion polymorphism of Siglec14 and its functional implications. Glycobiology 19:841–846.
39.
Landig CS, Hazel A, Kellman BP, Fong JJ, Schwarz F, Agarwal S, Varki N, Massari P, Lewis NE, Ram S, Varki A. 2019. Evolution of the exclusively human pathogen Neisseria gonorrhoeae: human-specific engagement of immunoregulatory Siglecs. Evol Appl 12:337–349.
40.
Sadarangani M, Pollard AJ, Gray-Owen SD. 2011. Opa proteins and CEACAMs: pathways of immune engagement for pathogenic Neisseria. FEMS Microbiol Rev 35:498–514.
41.
Gray-Owen SD, Dehio C, Haude A, Grunert F, Meyer TF. 1997. CD66 carcinoembryonic antigens mediate interactions between OPA-expressing Neisseria gonorrhoeae and human polymorphonuclear phagocytes. EMBO J 16:3435–3445.
42.
Alcott AM, Werner LM, Baiocco CM, Belcher Dufrisne M, Columbus L, Criss AK. 2022. Variable expression of Opa proteins by Neisseria gonorrhoeae influences bacterial association and phagocytic killing by human neutrophils. J Bacteriol 204:e0003522.
43.
Bos MP, Kao D, Hogan DM, Grant CCR, Belland RJ. 2002. Carcinoembryonic antigen family receptor recognition by gonococcal Opa proteins requires distinct combinations of hypervariable Opa protein domains. Infect Immun 70:1715–1723.
44.
Popp A, Dehio C, Grunert F, Meyer TF, Gray-Owen SD. 1999. Molecular analysis of Neisserial Opa protein interactions with the CEA family of receptors: identification of determinants contributing to the differential specificities of binding. Cell Microbiol 1:169–181.
45.
Smirnov A, Daily KP, Criss AK. 2014. Assembly of NADPH oxidase in human neutrophils is modulated by the opacity-associated protein expression state of Neisseria gonorrhoeae. Infect Immun 82:1036–1044.
46.
Sarantis H, Gray-Owen SD. 2007. The specific innate immune receptor CEACAM3 triggers neutrophil bactericidal activities via a Syk kinase-dependent pathway. Cell Microbiol 9:2167–2180.
47.
Buntru A, Roth A, Nyffenegger-Jann NJ, Hauck CR. 2012. HemITAM signaling by CEACAM 3, a human granulocyte receptor recognizing bacterial pathogens. Arch Biochem Biophys 524:77–83.
48.
Ball LM, Criss AK. 2013. Constitutively Opa-expressing and Opa-deficient Neisseria gonorrhoeae strains differentially stimulate and survive exposure to human neutrophils. J Bacteriol 195:2982–2990.
49.
Martin JN, Ball LM, Solomon TL, Dewald AH, Criss AK, Columbus L. 2016. Neisserial Opa protein-CEACAM interactions: competition for receptors as a means of bacterial invasion and pathogenesis. Biochemistry 55:4286–4294.
50.
John CM, Phillips NJ, Cardenas AJ, Criss AK, Jarvis GA. 2023. Comparison of lipooligosaccharides from human challenge strains of Neisseria gonorrhoeae. Front Microbiol 14:1215946.
51.
Mandrell RE, Schneider H, Apicella MA, Zollinger W, Rice PA, Griffiss JM. 1986. Antigenic and physical diversity of Neisseria gonorrhoeae lipooligosaccharides. Infect Immun 54:63–69.
52.
Wakarchuk WW, Gilbert M, Martin A, Wu Y, Brisson J, Thibault P, Richards JC. 1998. Structure of an alpha-2,6-sialylated lipooligosaccharide from Neisseria meningitidis immunotype L1 . European Journal of Biochemistry 254:626–633.
53.
Ram S, Gulati S, Lewis LA, Chakraborti S, Zheng B, DeOliveira RB, Reed GW, Cox AD, Li J, St Michael F, Stupak J, Su X-H, Saha S, Landig CS, Varki A, Rice PA. 2018. A novel sialylation site on Neisseria gonorrhoeae lipooligosaccharide links heptose II lactose expression with pathogenicity. Infect Immun 86:1617–1629.
54.
Wu ZL, Huang X, Burton AJ, Swift KAD. 2015. Glycoprotein labeling with click chemistry (GLCC) and carbohydrate detection. Carbohydr Res 412:1–6.
55.
Han Z, Thuy-Boun PS, Pfeiffer W, Vartabedian VF, Torkamani A, Teijaro JR, Wolan DW. 2021. Identification of an N-acetylneuraminic acid-presenting bacteria isolated from a human microbiome. Sci Rep 11:4763.
56.
Goon S, Schilling B, Tullius MV, Gibson BW, Bertozzi CR. 2003. Metabolic incorporation of unnatural sialic acids into Haemophilus ducreyi lipooligosaccharides. Proc Natl Acad Sci USA 100:3089–3094.
57.
Feigman MS, Kim S, Pidgeon SE, Yu Y, Ongwae GM, Patel DS, Regen S, Im W, Pires MM. 2018. Synthetic immunotherapeutics against gram-negative pathogens. Cell Chem Biol 25:1185–1194.
58.
Casey SG, Shafer WM, Spitznagel JK. 1986. Neisseria gonorrhoeae survive intraleukocytic oxygen-independent antimicrobial capacities of anaerobic and aerobic granulocytes in the presence of pyocin lethal for extracellular gonococci. Infect Immun 52:384–389.
59.
Criss AK, Katz BZ, Seifert HS. 2009. Resistance of Neisseria gonorrhoeae to non-oxidative killing by adherent human polymorphonuclear leucocytes. Cell Microbiol 11:1074–1087.
60.
Seib KL, Simons MP, Wu HJ, McEwan AG, Nauseef WM, Apicella MA, Jennings MP. 2005. Investigation of oxidative stress defenses of Neisseria gonorrhoeae by using a human polymorphonuclear leukocyte survival assay. Infect Immun 73:5269–5272.
61.
Nguyen GT, Green ER, Mecsas J. 2017. Neutrophils to the ROScue: mechanisms of NADPH oxidase activation and bacterial resistance. Front Cell Infect Microbiol 7:373.
62.
Rest RF. 1979. Killing of Neisseria gonorrhoeae by human polymorphonuclear neutrophil granule extracts. Infect Immun 25:574–579.
63.
Johnson MB, Ball LM, Daily KP, Martin JN, Columbus L, Criss AK. 2015. Opa+ Neisseria gonorrhoeae exhibits reduced survival in human neutrophils via Src family kinase-mediated bacterial trafficking into mature phagolysosomes. Cell Microbiol 17:648–665.
64.
Shafer WM, Martin LE, Spitznagel JK. 1986. Late intraphagosomal hydrogen ion concentration favors the in vitro antimicrobial capacity of a 37-kilodalton cationic granule protein of human neutrophil granulocytes. Infect Immun 53:651–655.
65.
Johnson MB, Criss AK. 2013. Neisseria gonorrhoeae phagosomes delay fusion with primary granules to enhance bacterial survival inside human neutrophils. Cell Microbiol 15:1323–1340.
66.
Werner LM, Palmer A, Smirnov A, Belcher Dufrisne M, Columbus L, Criss AK. 2020. Imaging flow cytometry analysis of CEACAM binding to Opa-expressing Neisseria gonorrhoeae. Cytometry A 97:1081–1089.
67.
Rest RF, Frangipane JV. 1992. Growth of Neisseria gonorrhoeae in CMP-N-acetylneuraminic acid inhibits nonopsonic (opacity-associated outer membrane protein-mediated) interactions with human neutrophils. Infect Immun 60:989–997.
68.
Smirnov A, Solga MD, Lannigan J, Criss AK. 2020. Using imaging flow cytometry to quantify neutrophil phagocytosis. Methods Mol Biol 2087:127–140.
69.
Jones C, Virji M, Crocker PR. 2003. Recognition of sialylated meningococcal lipopolysaccharide by Siglecs expressed on myeloid cells leads to enhanced bacterial uptake. Mol Microbiol 49:1213–1225.
70.
Graustein AD, Horne DJ, Fong JJ, Schwarz F, Mefford HC, Peterson GJ, Wells RD, Musvosvi M, Shey M, Hanekom WA, Hatherill M, Scriba TJ, Thuong NTT, Mai NTH, Caws M, Bang ND, Dunstan SJ, Thwaites GE, Varki A, Angata T, Hawn TR. 2017. The SIGLEC14 null allele is associated with Mycobacterium tuberculosis- and BCG-induced clinical and immunologic outcomes. Tuberculosis (Edinb) 104:38–45.
71.
Kelm S, Pelz A, Schauer R, Filbin MT, Tang S, de Bellard ME, Schnaar RL, Mahoney JA, Hartnell A, Bradfield P. 1994. Sialoadhesin, myelin-associated glycoprotein and CD22 define a new family of sialic acid-dependent adhesion molecules of the immunoglobulin superfamily. Curr Biol 4:965–972.
72.
Werner LM, Criss AK. 2023. Diverse functions of C4B-binding protein in health and disease. J Immunol 211:1443–1449.
73.
Islam EA, Anipindi VC, Francis I, Shaik-Dasthagirisaheb Y, Xu S, Leung N, Sintsova A, Amin M, Kaushic C, Wetzler LM, Gray-Owen SD. 2018. Specific binding to differentially expressed human carcinoembryonic antigen-related cell adhesion molecules determines the outcome of Neisseria gonorrhoeae infections along the female reproductive tract. Infect Immun 86:e00092-18.
74.
Shell DM, Chiles L, Judd RC, Seal S, Rest RF. 2002. The Neisseria lipooligosaccharide-specific alpha-2,3-sialyltransferase is a surface-exposed outer membrane protein. Infect Immun 70:3744–3751.
75.
Jen F-C, Ketterer MR, Semchenko EA, Day CJ, Seib KL, Apicella MA, Jennings MP. 2021. The Lst sialyltransferase of Neisseria gonorrhoeae can transfer keto-deoxyoctanoate as the terminal sugar of lipooligosaccharide: a glyco-achilles heel that provides a new strategy for vaccines to prevent gonorrhea. mBio 12:e03666-20.
76.
Chou HH, Hayakawa T, Diaz S, Krings M, Indriati E, Leakey M, Paabo S, Satta Y, Takahata N, Varki A. 2002. Inactivation of CMP-N-acetylneuraminic acid hydroxylase occurred prior to brain expansion during human evolution. Proc Natl Acad Sci USA 99:11736–11741.
77.
Patel PV, Parsons NJ, Andrade JRC, Nairn CA, Tan EL, Goldner M, Cole JA, Smith H. 1988. White blood cells including polymorphonuclear phagocytes contain a factor which induces gonococcal resistance to complement-mediated serum killing. FEMS Microbiol Lett 50:173–176.
78.
Eckhardt M, Gotza B, Gerardy-Schahn R. 1999. Membrane topology of the mammalian CMP-sialic acid transporter. J Biol Chem 274:8779–8787.
79.
Cross AS, Wright DG. 1991. Mobilization of sialidase from intracellular stores to the surface of human neutrophils and its role in stimulated adhesion responses of these cells. J Clin Invest 88:2067–2076.
80.
Rifat S, Kang TJ, Mann D, Zhang L, Puche AC, Stamatos NM, Goldblum SE, Brossmer R, Cross AS. 2008. Expression of sialyltransferase activity on intact human neutrophils. J Leukoc Biol 84:1075–1081.
81.
Sakarya S, Rifat S, Zhou J, Bannerman DD, Stamatos NM, Cross AS, Goldblum SE. 2004. Mobilization of neutrophil sialidase activity desialylates the pulmonary vascular endothelial surface and increases resting neutrophil adhesion to and migration across the endothelium. Glycobiology 14:481–494.
82.
Hong V, Steinmetz NF, Manchester M, Finn MG. 2010. Labeling live cells by copper-catalyzed alkyne--azide click chemistry. Bioconjug Chem 21:1912–1916.
83.
Ketterer MR, Rice PA, Gulati S, Kiel S, Byerly L, Fortenberry JD, Soper DE, Apicella MA. 2016. Desialylation of Neisseria gonorrhoeae lipooligosaccharide by cervicovaginal microbiome sialidases: the potential for enhancing infectivity in men. J Infect Dis 214:1621–1628.
84.
Gulati S, Schoenhofen IC, Lindhout-Djukic T, Lewis LA, Moustafa IY, Saha S, Zheng B, Nowak N, Rice PA, Varki A, Ram S. 2020. Efficacy of antigonococcal CMP-nonulosonate therapeutics require cathelicidins. J Infect Dis 222:1641–1650.
85.
Kellogg DS, Peacock WL, Deacon WE, Brown L, Pirkle DI. 1963. Neisseria gonorrhoeae. I. virulence genetically genetically linked to clonal variation. J Bacteriol 85:1274–1279.
86.
Ragland SA, Criss AK. 2019. Protocols to interrogate the interactions between Neisseria gonorrhoeae and primary human neutrophils. Methods Mol Biol 1997:319–345.
87.
Gulati S, Rice PA, Ram S. 2019. Complement-dependent serum bactericidal assays for Neisseria gonorrhoeae. Methods Mol Biol 1997:267–280.
88.
Achtman M, Neibert M, Crowe BA, Strittmatter W, Kusecek B, Weyse E, Walsh MJ, Slawig B, Morelli G, Moll A. 1988. Purification and characterization of eight class 5 outer membrane protein variants from a clone of Neisseria meningitidis serogroup A. J Exp Med 168:507–525.
89.
Geslewitz WE, Cardenas AJ, Zhou X, Zhang Y, Criss AK, Seifert HS. 2024. Development and implementation of a type I-C CRISPR-based programmable repression system for Neisseria gonorrhoeae. mBio 15:e0302523.
90.
Ferraro NJ, Kim S, Im W, Pires MM. 2021. Systematic assessment of accessibility to the surface of Staphylococcus aureus. ACS Chem Biol 16:2527–2536.
91.
Morse SA, Bartenstein L. 1980. Purine metabolism in Neisseria gonorrhoeae: the requirement for hypoxanthine. Can J Microbiol 26:13–20.
92.
Werner LM, Alcott A, Mohlin F, Ray JC, Belcher Dufrisne M, Smirnov A, Columbus L, Blom AM, Criss AK. 2023. Neisseria gonorrhoeae co-opts C4b-binding protein to enhance complement-independent survival from neutrophils. PLoS Pathog 19:e1011055.

Information & Contributors

Information

Published In

cover image mBio
mBio
Volume 15Number 58 May 2024
eLocator: e00119-24
Editor: M. Stephen Trent, University of Georgia, Athens, Georgia, USA
PubMed: 38587424

History

Received: 14 January 2024
Accepted: 12 March 2024
Published online: 9 April 2024

Keywords

  1. Neisseria gonorrhoeae
  2. gonorrhea
  3. lipooligosaccharide
  4. sialylation
  5. neutrophils
  6. infection
  7. reactive oxygen species
  8. degranulation

Contributors

Authors

Amaris J. Cardenas
Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
Author Contributions: Conceptualization, Formal analysis, Funding acquisition, Investigation, Methodology, Visualization, Writing – original draft, and Writing – review and editing.
Keena S. Thomas
Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
Author Contributions: Formal analysis, Investigation, and Writing – review and editing.
Mary W. Broden
Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
Author Contributions: Investigation and Writing – review and editing.
Noel J. Ferraro
Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
Author Contributions: Formal analysis, Investigation, Methodology, and Writing – review and editing.
Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
Author Contributions: Funding acquisition, Methodology, Supervision, and Writing – review and editing.
Constance M. John
VA Medical Center and University of California, San Francisco, San Francisco, California, USA
Author Contributions: Investigation, Methodology, and Writing – review and editing.
VA Medical Center and University of California, San Francisco, San Francisco, California, USA
Author Contributions: Funding acquisition, Supervision, and Writing – review and editing.
Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
Author Contributions: Conceptualization, Funding acquisition, Project administration, Supervision, Writing – original draft, and Writing – review and editing.

Editor

M. Stephen Trent
Editor
University of Georgia, Athens, Georgia, USA

Notes

The authors declare no conflict of interest.

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy