Bacterial pathogens are internalized into non-phagocytic cells either by a zipper mechanism involving a direct contact between a bacterial ligand and a cellular receptor or a trigger mechanism secondary to the formation of membrane ruffles. Here we show that internalization of capsulated Neisseria meningitidis within endothelial cells following type IV pilus-mediated adhesion is associated with the formation of cellular protrusions at the site of bacterial attachment. These protrusions, like microvilli, are highly enriched in ezrin and moesin, two members of the ERM(ezrin/radixin/moesin) family, whereas vinculin and paxillin are absent. ERM-binding transmembrane proteins, such as CD44, and cortical actin polymerization colocalized within these membrane protrusions. Expression of dominant-negative ezrin largely prevented cortical actin polymerization, thus confirming the role of this molecule in bacteria-induced cytoskeletal modifications. Moreover, using selective inhibitors and dominant-negative mutants of the Rho family GTPases, we show that bacteria-induced actin polymerization required the activation of both Rho and Cdc42 but not of Rac1. Whereas GTPase inhibition dramatically reduced actin polymerization at the site of bacterial attachment, ezrin recruitment was not affected, indicating that bacterial adhesion promotes ezrin recruitment independently of the activity of the Rho-GTPases. Furthermore, GTPase inhibition largely reduced N. meningitidis entry into endothelial cells without affecting adhesion. We thus propose that following pilus-mediated adhesion, capsulated N. meningitidis recruit ERM-binding transmembrane proteins, as well as ezrin and moesin, and that both Rho and Cdc42 are critical for the subsequent cytoskeletal modifications responsible for the formation of microvilli-like cellular protrusions and bacterial internalization.

The internalization of bacterial pathogens into non-phagocytic cells has been increasingly investigated and recognized as playing an essential role in bacteria—host-cell interactions. The first mode of entry is illustrated by Listeria and Yersinia, and it involves the formation of cell protrusions in tight contact with the bacterial surface. The second one is exemplified by Shigella and Salmonella and involves dramatic cytoskeletal rearrangements at the site of entry, with the formation of large membrane folds or ruffles around bacteria(Galan and Zhou, 2000;Nhieu and Sansonetti, 1999). Cytoskeletal modifications are known to require the activation of Rho family GTPases. Rho isoforms control the formation of actin stress fibers and focal adhesion plaques (Amano et al.,1997); Rac isoforms promote the formation of ruffling lamellipodia, and Cdc42 generates filopodia and microvilli structures(Gauthier-Rouviere et al.,1998; Hall, 1998). Whereas Shigella entry required Cdc42, Rac and Rho GTPases, invasion by Salmonella was only dependent upon Cdc42(Galan and Zhou, 2000;Nhieu and Sansonetti,1999).

Neisseria meningitidis (also referred to as meningococcus) is an extracellular pathogen that, once in the bloodstream, invades the meninges and the cerebrospinal fluid. A key event in N. meningitidis invasion is the interaction of bacteria with brain endothelial cells. Indeed, examination of post-mortem material has clearly shown a direct interaction of extracellular N. meningitidis with endothelial cells of both the choroid plexus and meningeal capillaries(Pron et al., 1997). Two meningococcal attributes are clearly essential for meningeal invasion: (i) the capsular polysaccharide, which allows bacterial survival in extracellular fluids, and (ii) type IV pili (TFP), which are multimeric structures essential for adhesion of virulent capsulated N. meningitidis to host cells(Nassif et al., 1999). This latter process seems to be due to a tip-located adhesin designated PilC(Rudel et al., 1995). In vitro studies using tight monolayers of human epithelial cells have shown that piliated capsulated meningococci can cross the monolayers by transcytosis(Merz et al., 1996;Pujol et al., 1997), thus suggesting that, in vivo, meningococci may use the transcellular route to cross the vascular endothelium to the meninges. For non-capsulated meningococci, as well as for the closely related pathogen N. gonorrhoeae, several mechanisms of bacterial internalization have been described, that involve interactions of cellular receptors with bacterial surface components, such as Opa and Opc outer membrane proteins(Bauer et al., 1999;Edwards et al., 2000;Meyer, 1998;Naumann et al., 1999;Virji et al., 1993;Virji et al., 1992;Virji et al., 1996). The role of these components in virulent capsulated strains of N. meningitidisis uncertain since non-piliated capsulated isolates are unable to efficiently interact with the cells. In addition, piliated capsulated N. meningitidis, which do not express Opa and Opc outer membrane proteins,can efficiently be internalized (Merz et al., 1996; Pujol et al.,1997; Pujol et al.,1999). Type IV pili thus remain the only component able to initiate the interaction of virulent capsulated N. meningitidis with human cells.

Pilus-mediated adhesion of capsulated strains was shown to lead to the formation of cortical plaques underneath bacterial colonies on the apical surface of epithelial cells. These structures result from the localized polymerization of cortical actin associated with the clustering of tyrosine phosphorylated and integral membrane proteins (ICAM-1, CD44, EGF receptor) as well as ezrin, a member of the ERM (ezrin-radixin-moesin) protein family(Merz et al., 1999;Merz and So, 1997). The role and the mechanisms by which these cytoskeletal modifications eventually lead to bacterial internalization remained mostly unknown. Recently we have observed in endothelial cells that ErbB2, a receptor-tyrosine kinase of the EGF receptor family, is specifically recruited underneath bacterial colonies following pilus-mediated adhesion. ErbB2 is activated by homodimer formation and is required for an efficient bacterial internalization, whereas ErbB2-associated signaling is not involved in bacteria-induced cytoskeletal modifications (Hoffmann et al.,2001).

The ERM proteins play a major structural and regulatory role in many of the morphogenic changes of the plasma membrane, including the formation of microvilli (Yonemura and Tsukita,1999). These proteins control the organization of the cortical cytoskeleton by acting as linkers between the plasma membrane and the actin cytoskeleton. ERM proteins interact through their amino-terminal domain with the cytoplasmic domain of transmembrane proteins (so-called ERM binding proteins), such as CD44 or ICAM-1, and interact with F-actin by their C-terminal domain. ERM proteins are in a cytosolic dormant form, in which the binding site for F-actin is masked by an intramolecular interaction between the N- and C-terminal domains (Mangeat et al., 1999). Activation of ERM seems to require the binding of phosphotidylinositol 4,5-biphosphate and the phosphorylation of a specific threonine residue in their C-terminal domain by Rho-kinase, an effector of Rho GTPase (Gautreau et al., 2000;Matsui et al., 1998;Matsui et al., 1999).

In this work, we show that the internalization of piliated capsulated meningococci in endothelial cells, a process associated with meningeal invasion, is triggered by the formation of microvilli-like membrane protrusions, but not ruffles, containing ezrin, moesin and the ERM-binding proteins CD44 and ICAM-1. The role of ezrin recruitment in actin polymerization was confirmed by expression of a dominant-negative form of ezrin. Furthermore, we provide evidence that both Rho and Cdc42, but not Rac1,are critical for actin cytoskeletal modifications induced by N. meningitidis and subsequent bacterial internalization, although ERM binding to the plasma membrane occurs through a Rho-GTPase-independent pathway. All together, these data indicate that the formation of microvilli-like membrane protrusions at the surface of human endothelial cells is critically involved in the internalization of piliated capsulated virulent N. meningitidis.

Bacterial strains

The strain used in this study, designated ROU (group W135, ET37), is a piliated capsulated isolate obtained from the cerebrospinal fluid of a 2-month-old infant and has been previously described(Pron et al., 1997). Strains were routinely grown on gonococcal base medium (GCB containing 4% medium base,1% agar, 0.4% glucose, 0.2 mg/ml thiamine, 0.0005%(Fe(NO3)3.9H2O, 0.01% L-glutamine) at 37°C under 5%CO2. The PilC- derivative of this isolate, which is incapable of pilus-mediated adhesion, has been previously described(Pron et al., 1997).

Cell culture and infection

Human umbilical vein endothelial cells (HUVECs; PromoCell, Heidelberg,Germany) were used between passages 1 and 8 and grown in Endo-SFM supplemented with 10% heat-inactivated FCS and 2 mM L-Glutamine (Life-Technologies, Grand Island, USA), 1 ng/ml of bFGF (Boehringer-Mannheim, Meylan, France), 0.5 UI/ml of heparin and 1.25 μg/ml of endothelial cell growth supplement (Sigma,Saint Louis, USA). The Human bone marrow endothelial cell line (HBMECs)(Schweitzer et al., 2000) was kindly provided by B. Weksler (Weill Medical College of Cornell University,NY, USA). Cells were cultured in DMEM-glutamax (Life-Technologies, Grand Island, USA) supplemented with 10% heat inactivated FCS, 7 UI/ml of heparin,7.5 μg/ml of endothelial cell growth supplement and 10 mM Hepes. Cells were seeded at 5×104 cells/cm2 in a 24-well or 6-well culture plates and grown at 37°C in a humidified incubator under 5%CO2 for 2 to 3 days.

The day before the infection, the culture medium was replaced by a serum-free medium (starvation medium). Approximately 107 bacteria in culture medium were added to the cells and allowed to adhere for 15-30 minutes. The monolayers were then washed every hour and new medium was added to avoid reinfection from the supernatant. At the indicated times, monolayers were harvested and the number of adhesive bacteria determined.

The number of internalized bacteria was determined by a gentamicin protection assay. Cells grown in a 24-well or 6-well culture plate were infected as above. 4 and 7 hours after infection, cells were washed and 150μg/ml of gentamicin was added to each well to kill extracellular bacteria. After 1 hour of incubation at 37°C, cells were scraped off the plates, and the number of intracellular bacteria was then determined. The proportion of internalized bacteria was calculated as the ratio of those that were gentamicin-resistant to adherent bacteria.

Where indicated, cells were pretreated with the following compounds,diluted in starvation medium: 1 ng/ml of Clostridium difficile toxin B (ToxB), a non-selective inhibitor of the Rho family GTPases(Boquet, 1999), kindly provided by P. Boquet (INSERM, Nice, France) or 30 μM of Y27632, a selective inhibitor of Rho kinase (Maekawa et al.,1999), an effector of the Rho protein, a gift from Yoshitomi Pharmaceutical (Osaka, Japan). These treatments were initiated 16 hours or 1 hour respectively, prior to the addition of bacteria and were maintained during the course of the experiments.

Antibodies

Anti-human CD44 mAb was purchased from R&D (Abington, UK), anti-ICAM-1,anti-VE-cadherin mAbs were from Serotec (Oxford, England), anti-paxillin mAb was from Transduction Laboratories (Lexington, USA) and anti-human vinculin mAb from Sigma (Saint Louis, USA). Phalloidin-alexa488 (Molecular Probes,Eugene, Or) was used to stain the actin cytoskeleton (F-actin). ERM proteins were detected using selective rabbit polyclonal antisera kindly provided by P. Mangeat (CNRS, Montpellier, France). mAb 26C4 specifically raised against RhoA(Lang et al., 1993) is a gift from J. Bertoglio (INSERM, Chatenay-Malabry, France). In infected monolayers,bacteria were stained using either ethidium bromide or a rabbit polyclonal antibody directed against the meningococcus strain Rou. This antibody was generated as follows: an overnight culture of Rou was either fixed with 2.5%paraformaldehyde or sonicated. These preparations were mixed with Freund's complete adjuvant (1:1), and 1 ml was used for inoculation of a New Zealand white rabbit. The rabbit was boosted with the same mixture three times every 2 weeks. Microinjected proteins were detected by immunostaining using antimyc 9E10 mAb from Biomol (Plymouth, USA) or anti-VSVG P5D4 mAb from Roche(Indianapolis, USA).

Immunofluorescence protocol and confocal microscopy

HUVECs were plated at a density of 5×104cells/cm2 onto 12-mm diameter acid-washed glass coverslips coated overnight with fibronectin (10 μg/ml). Cells were infected as described above, fixed with 2.5% paraformaldehyde in PBS for 20 minutes, neutralized with 0.1 M glycine in PBS for 5 minutes, permeabilized for 1 minute with 0.5%Triton X100 in PBS and then saturated for 20 minutes with PBS containing 0.2%gelatin before incubation for 30 minutes with the primary antibodies diluted in PBS/gelatin. Cells were then washed with PBS and incubated for 30 minutes with anti-rabbit or anti-mouse secondary antibodies conjugated with Cy3 or Cy5 and immunoabsorbed against human, bovine, rabbit or mouse serum proteins(Jackson Immunoresearch, West Grove, USA). Finally, cells were washed three times in PBS and mounted in moviol (Sigma, Saint Louis, USA) before analysis with a Zeiss LSM 510 confocal microscope. The pinhole of each PMT was adjusted separately in order to take XY optical sections with the same step. Sequential scannings were also performed in order to confirm the absence of any interference between signals detected at different wavelengths. Series were subjected to an orthogonal projection in order to make a two-dimensional reconstruction of the area under study.

Microinjection

Microinjections were performed using an Eppendorf Transjector 5246 coupled to an Eppendorf Micromanipulator 5179. HUVECs were seeded onto glass coverslips as described for the immunofluorescence protocol. About 50 cells per well were microinjected into the nucleus with the eukaryotic expression vector. The vector used was pRK5-myc, encoding the dominant-negative forms of Rac or Cdc42, RacN17 or Cdc42N17(Nobes and Hall, 1999). These plasmids were kindly provided by G.Tran Vhan Nieu (Institut Pasteur, Paris). pRK5-myc-RacN17 was microinjected at 117 μg/ml 18 hours before cell infection and pRK5-myc-Cdc42N17 at 133 μg/ml 5 hours before cell infection. Eukaryotic expression vectors encoding either the N- terminal domain of human ezrin (amino acids 1-309), a dominant-negative form of ezrin(pcB6-ezrin-Nter) or the full-length ezrin (pCB6-Ezrin) were obtained from A. Gautreau (Institut Curie, Paris). They were microinjected into the nuclei of HUVECs at a concentration of 120 μg/ml 2 hours prior infection of the monolayers with N. meningitidis.

The recombinant C3-transferase from Clostridium botulinum(Boquet, 1999), a Rho-selective inhibitor generously provided by M. Popoff (Institut Pasteur, Paris) was injected into the cytoplasm at 200 ng/ml with 2.5 mg/ml rhodamine-labelled dextran diluted in distilled water, 2 hours before infection by N. meningitidis as described above. Control injections carried out with rhodamine-labelled dextran did not produce any significant effect on cell morphology or actin organization.

Electron microscopy

For scanning electron microscopy, cells were cultured and infected as described above, then fixed with 2.5% glutaraldehyde solution in 0.1 M cacodylate buffer pH 7.3 Preparations were then coated with gold palladium after critical-point drying. The examination was performed on a JEOL 840A at the Centre Inter-Universitaire de Microscopie Electronique (Paris,France).

For transmission electron microscopy, cells were grown on transwells as described above and were fixed overnight at 4°C with a 1:1 mixture of 2.5%glutaraldehyde and 2.5% paraformaldehyde in cacodylate sucrose buffer (0.1 M cacodylate, 0.1 M sucrose, 5 mM CaCl2, 5 mM MgCl2, pH 7.2). Monolayers were then stained for 1 hour in a solution of 1%OsO4 and placed for 1 hour in 1% uranyl acetate. After dehydration in a graded series of alcohols, cells were embedded with polyester filter in Epon. Thin sections were obtained by using an Ultracut ultramicrotome and analyzed with JEOL-100CX electron microscope.

Transient formation of cellular protrusions is associated with the internalization of piliated capsulated meningococci in endothelial cells

In situ observations have demonstrated that meningococcal interactions with endothelial cells are a key event in the crossing of brain microvessels in the meninges and choroid plexuses (Pron et al., 1997). We therefore analyzed the interaction of N. meningitidis with human endothelial cells of both macrovascular and microvascular origin (HUVECs and HBMECs, respectively). The strain used in this work was a piliated capsulated isolate of ROU, obtained from the cerebrospinal fluid of a patient with fulminant meningococcal infection. Scanning electron microscopies are shown inFig. 1. As described for the interaction of N. meningitidis with epithelial cells(Pujol et al., 1997;Pujol et al., 1999), a two-step process was observed with bacteria interacting with endothelial monolayers. Following the initial attachment, bacteria proliferate and form small colonies, a step which is referred to as `localized adhesion'(Fig. 1A). Bacteria then disperse on the apical surface of the cells, forming a single monolayer covering the cells, a step referred to as `diffuse adhesion'(Fig. 1C). As expected, neither an interaction nor bacterial proliferation was observed with a PilC- capsulated derivative of ROU, which is incapable of pilus-mediated adhesion (data not shown).

Fig. 1.

Neisseria meningitidis induces localized cellular membrane protrusions. SEM examination of HUVEC monolayers infected with the ROU strain of N. meningitidis. Bars represent 1μm. (A) After 4 hours of infection, adhering bacteria proliferate and form small colonies at their site of attachment (localized adhesion). During this step, small protrusions of the cellular membrane are induced underneath bacterial colonies. (B) Detail showing cellular protrusions engulfing a bacterium during the localized adhesion step of the infection. (C) After 6 hours, meningococci start to spread over the apical cell surface (diffuse adhesion). (D) Higher magnification showing that the membrane protrusions have disappeared at the late stage of infection.

Fig. 1.

Neisseria meningitidis induces localized cellular membrane protrusions. SEM examination of HUVEC monolayers infected with the ROU strain of N. meningitidis. Bars represent 1μm. (A) After 4 hours of infection, adhering bacteria proliferate and form small colonies at their site of attachment (localized adhesion). During this step, small protrusions of the cellular membrane are induced underneath bacterial colonies. (B) Detail showing cellular protrusions engulfing a bacterium during the localized adhesion step of the infection. (C) After 6 hours, meningococci start to spread over the apical cell surface (diffuse adhesion). (D) Higher magnification showing that the membrane protrusions have disappeared at the late stage of infection.

Prior to bacterial infection, no membrane protrusion was observed on endothelial cells, whereas during localized adhesion of N. meningitidis, small protrusions of the cellular membranes were induced beneath and around bacteria of most colonies(Fig. 1A,B). The formation of these membrane protrusions was transient and disappeared during the stage of diffuse adhesion (Fig. 1C,D). At this later time point, even though bacteria remained highly adhesive to the endothelial cell surface, no intimate adhesion with pedestal formation was observed with endothelial cells, in contrast to our previous observations with infected epithelial cells (Pujol et al.,1999). Moreover, as shown inFig. 1, when interacting with endothelial cells, meningococci remained mostly extracellular. However, in some cases, cellular protrusions can be seen surrounding and engulfing meningococci (Fig. 1B). Transmission electron microscopy analysis(Fig. 2A-D) confirmed that bacteria can be found surrounded by such membrane protrusions, leading to the formation of large vacuoles containing meningococci. These observations strongly suggest that these protrusions are responsible for meningococcal internalization

Fig. 2.

Cellular membrane protrusions induced by N. meningitidis lead to bacteria engulfment within host cells. (A-D) TEM photographs of a HUVEC monolayer infected by N. meningitidis for 4 hours (localized adhesion). A,B,C show cellular protrusions engulfing bacteria. Bars represent 1 μm.

Fig. 2.

Cellular membrane protrusions induced by N. meningitidis lead to bacteria engulfment within host cells. (A-D) TEM photographs of a HUVEC monolayer infected by N. meningitidis for 4 hours (localized adhesion). A,B,C show cellular protrusions engulfing bacteria. Bars represent 1 μm.

To confirm that bacterial internalization is correlated with the formation of these cellular protrusions induced during the localized adhesion of N. meningitidis, gentamicin protection assays were performed at 4 hours(localized adhesion) and 8 hours (diffuse adhesion) after infection of the endothelial monolayers. As shown in Fig. 3, whereas the number of adhering bacteria dramatically increased between 4 hours and 8 hours, owing to extracellular proliferation, the number of internalized bacteria remained relatively unchanged during the same time,thus indicating that bacterial internalization predominantly takes place during the localized adhesion, when cellular protrusions are observed. Taken together, these observations suggest that piliated capsulated meningococci induce the transient formation of cellular protrusions during the initial adhesion phase of N. meningitidis infection and that these cytoskeletal modifications are associated with the internalization of a small fraction of adhesive bacteria. Ezrin is required for the assembly of the microvilli-like cellular protrusions

Fig. 3.

Adhesion versus internalization of N. meningitidis. Approximately 107 bacteria (ROU strain of N. meningitidis) in culture medium were added to the HUVECs and allowed to adhere for 15-30 minutes. The medium was changed every hour to avoid reinfection from the supernatant. At the indicated times (4 hours or 8 hours), monolayers were harvested and adherent bacteria counted. The number of internalized bacteria was determined by gentamicin protection assay, as described in Materials and Methods. Results are in Log CFU/ml.

Fig. 3.

Adhesion versus internalization of N. meningitidis. Approximately 107 bacteria (ROU strain of N. meningitidis) in culture medium were added to the HUVECs and allowed to adhere for 15-30 minutes. The medium was changed every hour to avoid reinfection from the supernatant. At the indicated times (4 hours or 8 hours), monolayers were harvested and adherent bacteria counted. The number of internalized bacteria was determined by gentamicin protection assay, as described in Materials and Methods. Results are in Log CFU/ml.

In order to gain insight into the molecular mechanisms responsible for the formation of the cellular protrusions induced by pilus-mediated adhesion of N. meningitidis, we identified components involved in their organization. Since the cortical actin-based cytoskeleton plays a crucial role in local membrane reorganization, we assessed, by immunofluorescence analysis,whether actin was polymerized in the membrane protrusions induced by N. meningitidis. As shown in Fig. 4A, localized polymerization of cortical actin was observed underneath bacterial colonies. Moreover, xz section analysis of the actin staining (Fig. 4D) revealed that cortical actin was indeed polymerized in the membrane protrusions.

Fig. 4.

Cellular protrusions induced by N. meningitidis are associated with ezrin recruitment and actin polymerization. Monolayers of HUVECs were infected for 4 hours with the ROU strain of N. meningitidis. A,B,C:xy sections performed by confocal microscopy of double fluorescence labeling((A) bacteria and F-actin) or triple fluorescence labeling ((B) bacteria,F-actin and paxillin staining; (C) F-actin, ezrin and CD44) in the same fields. (D) xz section of a double fluorescence labeling of F-actin and ezrin.(A,C,D): ×200; (B) ×63.

Fig. 4.

Cellular protrusions induced by N. meningitidis are associated with ezrin recruitment and actin polymerization. Monolayers of HUVECs were infected for 4 hours with the ROU strain of N. meningitidis. A,B,C:xy sections performed by confocal microscopy of double fluorescence labeling((A) bacteria and F-actin) or triple fluorescence labeling ((B) bacteria,F-actin and paxillin staining; (C) F-actin, ezrin and CD44) in the same fields. (D) xz section of a double fluorescence labeling of F-actin and ezrin.(A,C,D): ×200; (B) ×63.

We then analyzed the distribution of various proteins known to link the actin cytoskeleton and membrane components. Paxillin(Fig. 4B) and vinculin (data not shown) were not recruited to the site of polymerization of cortical actin beneath bacterial colonies but remained highly enriched at the tip of actin stress fibers in focal adhesions, indicating that cytoskeletal modifications induced by N. meningitidis differed from focal adhesions or focal complexes that are seen in ruffles (Yamada and Geiger, 1997; Zamir et al., 1999). On the other hand, using specific antibodies, ezrin(Fig. 4C) and moesin (not shown) were found to be recruited underneath bacterial colonies to the site of actin polymerization together with the ERM-binding proteins CD44(Fig. 4C) and ICAM-1 (not shown), whereas radixin, another member of the ERM family, remained localized at cell junctions (not shown). In addition, xz section analysis of ezrin distribution in infected cells showed a massive recruitment just beneath the plasma membrane at the tip of the cellular protrusions(Fig. 4D), reminiscent of ezrin distribution in epithelial microvilli(Yonemura and Tsukita,1999).

The ERM N-terminal domain interacts with ERM-binding proteins whereas the C-terminal domain binds to F-actin. Indeed, the N-terminal domain (amino acids 1-309) of ezrin can compete with ezrin for interaction with ERM-binding proteins, thus behaving as a dominant-negative form of ezrin. In order to assess the actual function of ezrin in the assembly of the bacteria-induced cellular protrusions, we expressed its N-terminal domain in endothelial cells before N. meningitidis infection. As shownFig. 5A, expression of this dominant-negative form of ezrin not only inhibited the recruitment of endogenous ezrin but also prevented polymerization of actin underneath bacterial colonies. This observation was further confirmed by cell counting(Fig. 5B). It should be pointed out that microinjection of full-length ezrin had no effect on actin polymerization as shown in Fig. 5C, thus eliminating the possibility that the lack of actin polymerization observed in Fig. 5B was a consequence of microinjection. These data clearly indicate that ezrin is playing a pivotal role in the cytoskeletal modifications induced by pilus-mediated adhesion of N. meningitidis.

Fig. 5.

Ezrin recruitment is responsible for actin polymerization. (A) 2 hours prior to infection, HUVECs were microinjected with pCB6-Nter-Ezrin-VSVG,encoding the VSVG-tagged truncated N-terminal domain of ezrin, as a dominant-negative form of ezrin. Cells were infected with the ROU strain of N. meningitidis for 4 hours and then labeled for F-actin, endogenous ezrin and VSVG-tag (Nter-ezrin). In the transfected cells a large amount of VSVG-labeled truncated ezrin was recruited below the bacterial colonies (top right) without actin polymerization (top left). Endogenous ezrin (bottom right) was recruited underneath bacterial colonies in non-transfected cells. Transfection with the dominant-negative form almost completely prevented the recruitment of endogenous ezrin. (B) Bacterial colonies with polymerized actin and/or recruited endogenous ezrin were counted by immunofluorescence analysis in non-transfected cells or cells transfected with a plasmid encoding the VSVG-tagged truncated amino-terminal domain of ezrin (pCB6-ezrin-Nter). These data correspond to the results of at least six independent experiments. Results are expressed in percent of adherent colonies. (C) Bacterial colonies with polymerized actin were counted by immunofluorescence analysis in HUVECs that were transfected with a plasmid encoding full length ezrin (pCB6-ezrin)or the VSVG-tagged truncated amino-terminal domain of ezrin (pCB6-ezrin-Nter). Results are expressed in percent of adherent colonies.

Fig. 5.

Ezrin recruitment is responsible for actin polymerization. (A) 2 hours prior to infection, HUVECs were microinjected with pCB6-Nter-Ezrin-VSVG,encoding the VSVG-tagged truncated N-terminal domain of ezrin, as a dominant-negative form of ezrin. Cells were infected with the ROU strain of N. meningitidis for 4 hours and then labeled for F-actin, endogenous ezrin and VSVG-tag (Nter-ezrin). In the transfected cells a large amount of VSVG-labeled truncated ezrin was recruited below the bacterial colonies (top right) without actin polymerization (top left). Endogenous ezrin (bottom right) was recruited underneath bacterial colonies in non-transfected cells. Transfection with the dominant-negative form almost completely prevented the recruitment of endogenous ezrin. (B) Bacterial colonies with polymerized actin and/or recruited endogenous ezrin were counted by immunofluorescence analysis in non-transfected cells or cells transfected with a plasmid encoding the VSVG-tagged truncated amino-terminal domain of ezrin (pCB6-ezrin-Nter). These data correspond to the results of at least six independent experiments. Results are expressed in percent of adherent colonies. (C) Bacterial colonies with polymerized actin were counted by immunofluorescence analysis in HUVECs that were transfected with a plasmid encoding full length ezrin (pCB6-ezrin)or the VSVG-tagged truncated amino-terminal domain of ezrin (pCB6-ezrin-Nter). Results are expressed in percent of adherent colonies.

Clostridium difficile toxin B inhibits cortical actin polymerization but not ezrin recruitment underneath bacterial colonies

To investigate the putative role of Rho family GTPases in the organization of membrane protrusions induced by N. meningitidis, endothelial cell monolayers were incubated with Clostridium difficile toxin B (Tox B),which non-selectively inhibits all members of the Rho family(Boquet, 1999). The number of bacterial colonies inducing cortical actin polymerization and/or ezrin recruitment was measured (Fig. 6A and 6B). Tox B induced a dramatic decrease in the number of colonies associated with cortical actin polymerization(Fig. 6B). However, the number of bacterial colonies recruiting ezrin was not affected(Fig. 6A), although, as shown in Fig. 6C, ezrin recruitment was less pronounced than that observed with no ToxB treatment when actin was polymerized. We then conclude that Rho family GTPases are involved in cortical actin polymerization but not in the initial ezrin and ezrin-binding protein recruitment induced by N. meningitidis underneath bacterial colonies.

Fig. 6.

Treatment by C. difficile toxin B prevents actin polymerization but not ezrin recruitment induced by N. meningitidis. (A,B) Confluent monolayers of HUVECs were either left untreated (control) or treated with 1 ng/ml of C. difficile toxin B (ToxB) in starvation medium for 16 hours prior to the infection. Control and Tox-B-treated cells were then infected with the ROU strain of N. meningitidis for 4 hours. Tox B treatment was maintained during infection. Cells were double stained for F-actin and ezrin and the percentage of bacterial colonies recruiting ezrin(A) or F-actin-positive (B) was determined by immunofluorescence analysis. Results correspond to counts of 300 bacterial colonies in three distinct experiments. (C) xy section of double fluorescence labeling of F-actin (middle panel) and ezrin (right panel) in the same field. Analysis was performed by confocal microscopy at magnification ×63. The localization of the bacterial colony in the same field is shown by phase contrast (left panel).

Fig. 6.

Treatment by C. difficile toxin B prevents actin polymerization but not ezrin recruitment induced by N. meningitidis. (A,B) Confluent monolayers of HUVECs were either left untreated (control) or treated with 1 ng/ml of C. difficile toxin B (ToxB) in starvation medium for 16 hours prior to the infection. Control and Tox-B-treated cells were then infected with the ROU strain of N. meningitidis for 4 hours. Tox B treatment was maintained during infection. Cells were double stained for F-actin and ezrin and the percentage of bacterial colonies recruiting ezrin(A) or F-actin-positive (B) was determined by immunofluorescence analysis. Results correspond to counts of 300 bacterial colonies in three distinct experiments. (C) xy section of double fluorescence labeling of F-actin (middle panel) and ezrin (right panel) in the same field. Analysis was performed by confocal microscopy at magnification ×63. The localization of the bacterial colony in the same field is shown by phase contrast (left panel).

Cdc42 and Rho are required for actin polymerization induced by N. meningitidis and bacterial internalization

We next determined which GTPases of the Rho family were involved in the cytoskeletal modifications induced by N. meningitidis. For that purpose, we used a monoclonal antibody specifically raised against RhoA, 26C4(Lang et al., 1997). Immunofluorescence analysis of RhoA distribution in infected cells demonstrated an enrichment of this molecule in the microvilli-like structures (Fig. 7A). Furthermore, the role of Rho was further analyzed using (i) Clostridium botulinum C3 transferase, a selective inhibitor of Rho,and (ii) Y27632, a selective inhibitor of Rho kinase, a Rho effector(Maekawa et al., 1999). Under the conditions used, Y27632 induced a loss of actin stress fibers, without inducing cell rounding, and prevented actin polymerization underneath bacterial colonies (Fig. 7B). Indeed, the number of bacterial colonies able to induce cortical actin polymerization was significantly lower in cells incubated with Y27632 than in control cells (Fig. 8). Similar results were obtained after microinjection of C3 transferase(Fig. 8). All together, these data demonstrate the role of Rho and of its effector Rho kinase in mediating cortical actin polymerization induced by N. meningitidis.

Fig. 7.

Effect of GTPase inhibition on actin polymerization induced by N. meningitidis. (A) HUVECs were infected for 4 hours with the ROU strain of N. meningitidis. Cells were labeled for bacteria, F-actin and RhoA,and xz section analysis was performed by confocal microscopy at magnification×300.

(B) HUVECs, pretreated with the Rho kinase inhibitor Y27632 at 30 μM in starvation medium for 1 hour, were infected for 4 hours in the presence of the inhibitor and stained for actin (right panel, xy section at magnification ×63). The localization of the bacterial colony in the same field is shown by Nomarsky (left panel).

(C) HUVECs were microinjected with the cDNA encoding the dominant-negative form of Cdc42 (Cdc42N17) coupled to a myc tag, 5 hours prior to cell infection. Cells were then labeled for bacteria, F-actin and the myc tag. xy sections were performed by confocal microscopy at magnification ×100. xz sections showed in the lower panels were performed along the indicated lines of the transfected cell and the non transfected cell as control at magnification ×100.

(D) HUVECs were microinjected with the cDNA encoding the dominant-negative form of Rac1 (Rac1N17) coupled to a myc tag, 18 hours prior to cell infection. Cells were then labeled for bacteria, actin and the myc tag. xy sections were performed by confocal microscopy at magnification ×63. The arrows in D localize the polymerized actin visible either on control cell or microinjected cell.

Fig. 7.

Effect of GTPase inhibition on actin polymerization induced by N. meningitidis. (A) HUVECs were infected for 4 hours with the ROU strain of N. meningitidis. Cells were labeled for bacteria, F-actin and RhoA,and xz section analysis was performed by confocal microscopy at magnification×300.

(B) HUVECs, pretreated with the Rho kinase inhibitor Y27632 at 30 μM in starvation medium for 1 hour, were infected for 4 hours in the presence of the inhibitor and stained for actin (right panel, xy section at magnification ×63). The localization of the bacterial colony in the same field is shown by Nomarsky (left panel).

(C) HUVECs were microinjected with the cDNA encoding the dominant-negative form of Cdc42 (Cdc42N17) coupled to a myc tag, 5 hours prior to cell infection. Cells were then labeled for bacteria, F-actin and the myc tag. xy sections were performed by confocal microscopy at magnification ×100. xz sections showed in the lower panels were performed along the indicated lines of the transfected cell and the non transfected cell as control at magnification ×100.

(D) HUVECs were microinjected with the cDNA encoding the dominant-negative form of Rac1 (Rac1N17) coupled to a myc tag, 18 hours prior to cell infection. Cells were then labeled for bacteria, actin and the myc tag. xy sections were performed by confocal microscopy at magnification ×63. The arrows in D localize the polymerized actin visible either on control cell or microinjected cell.

Fig. 8.

Rho and Cdc42, but not Rac 1, are involved in actin polymerization induced by N. meningitidis. Confluent monolayers of HUVECs were either left untreated (control), treated with 30 μM Y27632 prior to the infection and during the infection (Y27632), microinjected with the recombinant exoenzyme C3 of C. botulinum 2 hours before cell infection (C3) or microinjected with the cDNA encoding a dominant-negative form of Rac (RacN17) or Cdc42 (Cdc42N17) 18 hours or 5 hours prior to infection of the monolayer, respectively. Cells were then infected for 4 hours and labeled for bacteria, F-actin and myc tag. The percentages of bacterial colonies associated with polymerized actin were determined by immunofluorescence analysis. Each condition was compared with non-treated cells infected in the same conditions. Results correspond to counts of 1600 infected cells in 3 to 10 distinct experiments per condition.

Fig. 8.

Rho and Cdc42, but not Rac 1, are involved in actin polymerization induced by N. meningitidis. Confluent monolayers of HUVECs were either left untreated (control), treated with 30 μM Y27632 prior to the infection and during the infection (Y27632), microinjected with the recombinant exoenzyme C3 of C. botulinum 2 hours before cell infection (C3) or microinjected with the cDNA encoding a dominant-negative form of Rac (RacN17) or Cdc42 (Cdc42N17) 18 hours or 5 hours prior to infection of the monolayer, respectively. Cells were then infected for 4 hours and labeled for bacteria, F-actin and myc tag. The percentages of bacterial colonies associated with polymerized actin were determined by immunofluorescence analysis. Each condition was compared with non-treated cells infected in the same conditions. Results correspond to counts of 1600 infected cells in 3 to 10 distinct experiments per condition.

We next investigated whether the related proteins, Rac1 and Cdc42, also affected N. meningitidis-induced actin polymerization. HUVECs were microinjected with plasmids encoding Rac1N17 and Cdc42N17, the dominant-negative forms of Rac1 and Cdc42,respectively. As shown in Fig. 8, the number of bacterial colonies with polymerized actin was significantly reduced in endothelial cells microinjected with Cdc42N17, whereas microinjection of Rac1N17 had no effect. Fig. 7C shows that microinjection of Cdc42N17 induced a dramatic increase in the formation of stress fibers, a cytoskeletal organization controlled by Rho(Hall, 1998), without inducing polymerization of cortical actin underneath bacterial colonies (see xz section in Fig. 7C). This increase in the formation of stress fibers in cells microinjected with a dominant-negative form of Cdc42 indicates that the effect of Cdc42 N17 on actin cytoskeleton is not due to an inhibition of Rho activity.Fig. 7D confirmed that microinjection of the dominant-negative form of Rac1 had no effect on cortical actin polymerization induced by N. meningitidis, even though microinjected cells were retracted and had no stress fibers, as expected from Rac1 inhibition (Hall, 1998). Taken together, these data demonstrate that the cytoskeletal modifications induced by N. meningitidis required the activation of both Cdc42 and Rho but not of Rac1.

To confirm that bacterial internalization is promoted by the induction of these cytoskeleton changes, we analyzed the effect of ToxB and Y27632 on bacterial adhesion and entry into endothelial cells. As shown inFig. 9, bacterial entry into cells treated by either of these two inhibitors was significantly decreased,whereas N. meningitidis adhesion was unaffected. These data demonstrate that actin cytoskeleton reorganization induced by both Cdc42 and Rho, via Rho kinase activation, are crucial for the internalization of N. meningitidis into endothelial cells. They suggest that in vivo activation of both Cdc42 and Rho during pilus-mediated adhesion onto brain endothelial cells may be involved in meningeal invasion by N. meningitidis.

Fig. 9.

Rho GTPases are involved in N. meningitidis internalization into endothelial host cells. HBMECs were either left untreated (control) or pretreated for 18 hours with 1 ng/ml of C. difficile toxin B (ToxB)or for 2 hours with 30 μM Y27632 (Y27632). Cells were infected with the ROU strain of N. meningitidis for 4 hours in the presence of the inhibitors, and the number of adherent bacteria (A) and internalized bacteria(B) were determined as in Fig. 3. These experiments, carried out in triplicates, were performed four times independently and results from one representative experiment are shown.

Fig. 9.

Rho GTPases are involved in N. meningitidis internalization into endothelial host cells. HBMECs were either left untreated (control) or pretreated for 18 hours with 1 ng/ml of C. difficile toxin B (ToxB)or for 2 hours with 30 μM Y27632 (Y27632). Cells were infected with the ROU strain of N. meningitidis for 4 hours in the presence of the inhibitors, and the number of adherent bacteria (A) and internalized bacteria(B) were determined as in Fig. 3. These experiments, carried out in triplicates, were performed four times independently and results from one representative experiment are shown.

Few bacterial pathogens are capable of crossing the blood brain barrier. It is therefore likely that these neuroinvasive bacteria have developed sophisticated tools that enable them to do so, either by a paracellular or a transcellular route. In the case of Streptococcus pneumoniae,trafficking towards the cerebral tissue seems to occur by a transendothelial migration induced by the interaction between bacterial phosporylcholine and the endothelial receptor of platelet-activating factor(Cundell et al., 1996;Cundell et al., 1995;Ring et al., 1998). Previously reported in vivo data have clearly demonstrated that an important step in meningeal invasion by N. meningitidis is its interaction with brain endothelial cells. In this study, we present data that indicate that meningococci are internalized into vascular endothelial cells by triggering multiple signaling pathways within the host cells. We provide evidence that the cytoskeletal modifications associated with the internalization of N. meningitidis into endothelial cells are associated with the formation of cellular protrusions, which are reminiscent of epithelial microvilli in terms of shape and ezrin content. These structures are probably responsible for bacterial engulfment and internalization. On the basis of the capacity of an ezrin dominant-negative form to block actin polymerization, our data suggest that these events occur in a two-step process: first, the recruitment of ezrin and moesin underneath bacterial colonies; second, the polymerization of cortical actin. Moreover, the present study shows that the first step is not dependent on Rho GTPases, whereas the second step requires the activation of Rho, and its downstream effector Rho kinase, as well as Cdc42, but not Rac1.

The precise mechanism by which pilus-mediated adhesion is recruiting ezrin is still unknown. ERM proteins have been shown to be present in the cytoplasm as oligomers, which can be recruited to the cytoplasmic membrane where threonine phosphorylation in the C-terminal domain is required for the transition to monomers. Only monomers can crosslink actin filaments to the plasma membrane to form microvilli in epithelial cells(Gautreau et al., 2000). The N-terminal domain is responsible for membrane targeting via interaction with ezrin-binding proteins, such as CD44 or ICAM-1. Indeed, CD44 has been proposed to be involved in the formation of microvilli by concentrating activated ERM proteins at the plasma membrane (Yonemura et al., 1998; Yonemura and Tsukita, 1999). We therefore hypothesize that, during pilus-mediated adhesion of N. meningitidis, the recruitment of ezrin-binding proteins might similarly target ezrin and moesin to the plasma membrane, underneath bacterial colonies, where they would be activated and would trigger actin polymerization.

Several studies have shown that Rho stimulates the activity of ezrin and moesin (i) by inducing their recruitment to the plasma membrane, (ii) by promoting the formation of phosphotidylinositol 4,5-biphosphate, which is responsible for an activating conformational change in ERM proteins, and(iii), although still a matter of debate(Matsui et al., 1998;Matsui et al., 1999), by activating Rho kinase, which can phosphorylate ERM proteins in their C-terminal domain. Surprisingly, we observed in the present study that ToxB, a non-selective inhibitor of Rho family GTPases, did not affect the recruitment of ERM proteins or ERM-binding proteins underneath bacterial colonies. Further investigation will aim at identifying the alternative signaling pathways involved in ERM protein recruitment promoted by the pilus-mediated adhesion of N. meningitidis.

Our data using specific inhibitors of the Rho family GTPases demonstrate the crucial role of both Rho and Cdc42 in N. meningitidis-induced actin polymerization and bacterial entry. However, their respective contribution still remains unknown, as does the identity of Cdc42 targets. Considering the role of Rho kinase in actin polymerization induced by N. meningitidis, it is tempting to speculate that the role of Rho activation is to promote ezrin phosphorylation following its recruitment to the plasma membrane at the site of bacterial attachment. Since a role for Cdc42 in the formation of microvilli-like structures has previously been reported in epithelial cells (Gauthier-Rouviere et al.,1998), we hypothesize here that it might be responsible for actin nucleation leading to the formation of the microvilli-like membrane protrusions in endothelial cells.

Interestingly, the membrane protrusions induced by N. meningitidison the endothelial surface significantly differ from those induced by other known invasive bacteria. Cytoskeletal modifications leading to the formation of ruffles have been shown to be responsible for the internalization of bacterial pathogens such as Shigella, Salmonella and, more recently, Neisseria gonorrhoeae (Edwards et al., 2000; Galan and Zhou,2000; Nhieu and Sansonetti,1999). In the case of Shigella, ezrin recruitment was shown to be dependent on bacteria-induced actin polymerization(Skoudy et al., 1999). Moreover, Shigella-induced protrusions are enriched in paxillin and vinculin, two proteins usually localized at focal adhesions, which are not recruited in N. meningitidis-induced membrane protrusions. Indeed,the lack of ruffle formation reported in this study in response to N. meningitidis adhesion was correlated with our observation that Rac1,which generally controls ruffle formation(Hall, 1998), is not involved in bacteria-induced actin cytoskeleton modifications in endothelial cells.

Despite the lack of the type III secretion system, which is involved in Shigella and Salmonella internalization, virulent capsulated N. meningitidis are internalized into host cells. This internalization is likely to promote their subsequent transcytosis through cellular barriers in vivo. As previously mentioned, type IV pili are the only bacterial components identified so far involved in the formation of the microvilli-like membrane protrusions induced by N. meningitidis. That pilusmediated adhesion has been shown to be important for meningeal invasion(Pron et al., 1997) is consistent with the role of these cytoskeletal modifications in the crossing of the cerebral vascular endothelium. It is, however, possible that additional meningococcal components are involved in the signaling events leading to the formation of the membrane protrusions observed here. Recently, a role for the lipooligosaccharide (LOS) in the actin cytoskeleton rearrangement induced by piliated Neisseria gonorrhoeae has been shown(Song et al., 2000). Gonococcal strains expressing LOS lacking the lacto-N-neotetraose did not promote actin polymerization and were less invasive. However, we did not observe here such inhibition of actin polymerization with a mutant of the ROU strain lacking the LOS core (data not shown).

The present study thus provides new insights into the molecular mechanisms used by N. meningtidis to enter endothelial cells, a critical step in the pathogenesis of this bacteria for its trafficking through the cerebral vasculature. Further studies on the meninogoccal components and their cellular receptors involved in this interaction should provide essential clues to the understanding of the molecular mechanisms of N. meningitidisinfection of the central nervous system.

We thank Patrice Boquet, Guy Tran Van Nhieu, Paul Mangeat, Alexis Gautreau,and Michel Popoff for fruitful discussions and for providing us with materials, Joanne Dove for careful reading of the manuscript. X.N.'s laboratory is supported by INSERM, and the Université RenéDescartes ParisV. This work was supported in part by a special grant from the`Programme de Recherche Fondamentale en Microbiologie et Maladies Infectieuses'.

Amano, M., Chihara, K., Kimura, K., Fukata, Y., Nakamura, N.,Matsuura, Y. and Kaibuchi, K. (
1997
). Formation of actin stress fibers and focal adhesions enhanced by Rho-kinase.
Science
275
,
1308
-1311.
Bauer, F. J., Rudel, T., Stein, M. and Meyer, T. F.(
1999
). Mutagenesis of the Neisseria gonorrhoeae porin reduces invasion in epithelial cells and enhances phagocyte responsiveness.
Mol. Microbiol.
31
,
903
-913.
Boquet, P. (
1999
). Bacterial toxins inhibiting or activating small GTP-binding proteins.
Annals NY Acad. Sci.
886
,
83
-90.
Cundell, D. R., Gerard, C., Idanpaan-Heikkila, I., Tuomanen, E. I. and Gerard, N. P. (
1996
). PAf receptor anchors Streptococcus pneumoniae to activated human endothelial cells.
Adv. Exp. Med. Biol.
416
,
89
-94.
Cundell, D. R., Gerard, N. P., Gerard, C., Idanpaan-Heikkila, I. and Tuomanen, E. I. (
1995
). Streptococcus pneumoniaeanchor to activated human cells by the receptor for platelet-activating factor.
Nature
377
,
435
-438.
Edwards, J. L., Shao, J. Q., Ault, K. A. and Apicella, M. A.(
2000
). Neisseria gonorrhoeae elicits membrane ruffling and cytoskeletal rearrangements upon infection of primary human endocervical and ectocervical cells.
Infect. Immunity
68
,
5354
-5363.
Galan, J. E. and Zhou, D. (
2000
). Striking a balance: modulation of the actin cytoskeleton by Salmonella.
Proc. Natl. Acad. Sci. USA
97
,
8754
-8761.
Gauthier-Rouviere, C., Vignal, E., Meriane, M., Roux, P.,Montcourier, P. and Fort, P. (
1998
). RhoG GTPase controls a pathway that independently activates Rac1 and Cdc42Hs.
Mol. Biol. Cell
9
,
1379
-1394.
Gautreau, A., Louvard, D. and Arpin, M. (
2000
). Morphogenic effects of ezrin require a phosphorylation-induced transition from oligomers to monomers at the plasma membrane.
J. Cell Biol.
150
,
193
-203.
Hall, A. (
1998
). Rho GTPases and the actin cytoskeleton.
Science
279
,
509
-514.
Hoffmann, I., Eugène, E., Nassif, X., Couraud, P. C. and Bourdoulous, S. (
2001
). Activation of ErbB2 receptor tyrosine kinase supports invasion of endothelial cells by Neisseria meningitidis.
J. Cell Biol
.
155
,
133
-143.
Lang, P., Gesbert, F., Thiberge, J. M., Troalen, F., Dutartre,H., Chavrier, P. and Bertoglio, J. (
1993
). Characterization of a monoclonal antibody specific for the Ras-related GTP-binding protein Rho A.
Biochem. Biophys. Res. Commun.
196
,
1522
-1528.
Maekawa, M., Ishizaki, T., Boku, S., Watanabe, N., Fujita, A.,Iwamatsu, A., Obinata, T., Ohashi, K., Mizuno, K. and Narumiya, S.(
1999
). Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase.
Science
285
,
895
-898.
Mangeat, P., Roy, C. and Martin, M. (
1999
). ERM proteins in cell adhesion and membrane dynamics.
Trends Cell Biol.
9
,
187
-192.
Matsui, T., Maeda, M., Doi, Y., Yonemura, S., Amano, M.,Kaibuchi, K. and Tsukita, S. (
1998
). Rho-kinase phosphorylates COOH-terminal threonines of ezrin/radixin/moesin (ERM) proteins and regulates their head-to-tail association.
J. Cell Biol.
140
,
647
-657.
Matsui, T., Yonemura, S. and Tsukita, S.(
1999
). Activation of ERM proteins in vivo by Rho involves phosphatidyl-inositol 4-phosphate 5-kinase and not ROCK kinases.
Curr. Biol.
9
,
1259
-1262.
Merz, A. J., Rifenbery, D. B., Grove Arvidson, C. and So, M.(
1996
). Traversal of a polarized epithelium by pathogenic Neisseriae: facilitation by type IV pili and maintenance of epithelial barrier function.
Mol. Medicine
2
,
745
-754.
Merz, A. J. and So, M. (
1997
). Attachment of piliated, Opa(-) and Opc(-) gonococci and meningococci to epithelial cells elicits cortical actin rearrangements and clustering of tyrosine-phosphorylated proteins.
Infect. Immun.
65
,
4341
-4349.
Merz, A. J., Enns, C. A. and So, M. (
1999
). Type IV pili of pathogenic Neisseriae elicit cortical plaque formation in epithelial cells.
Mol. Microbiol.
32
,
1316
-1332.
Meyer, T. F. (
1998
). Pathogenic Neisseria—interplay between pro- and eukaryotic worlds.
Folia Microbiologica
43
,
311
-319.
Nassif, X., Pujol, C., Morand, P. and Eugene, E.(
1999
). Interactions of pathogenic Neisseria with host cells. Is it possible to assemble the puzzle?
Mol. Microbiol.
32
,
1124
-1132.
Naumann, M., Rudel, T. and Meyer, T. F. (
1999
). Host cell interactions and signalling with Neisseria gonorrhoeae.
Curr. Opin. Microbiol
.
2
,
62
-70.
Nhieu, G. T. and Sansonetti, P. J. (
1999
). Mechanism of Shigella entry into epithelial cells.
Curr. Opin. Microbiol.
2
,
51
-55.
Nobes, C. D. and Hall, A. (
1999
). Rho GTPases control polarity, protrusion, and adhesion during cell movement.
J. Cell Biol.
144
,
1235
-1244.
Pron, B., Taha, M.-K., Rambaud, C., Fournet, J.-C., Pattey, N.,Monnet, J.-P., Musilek, M., Beretti, J.-L. and Nassif, X.(
1997
). Interaction of Neisseria meningitidis with the components of the blood-brain barrier correlates with an increased expression of PilC.
J. Infect. Dis.
176
,
1285
-1292.
Pujol, C., Eugène, E., de Saint Martin, L. and Nassif,X. (
1997
). Interaction of Neisseria meningitidiswith a polarized monolayer of epithelial cells.
Infect. Immun.
65
,
4836
-4842.
Pujol, C., Eugene, E., Marceau, M. and Nassif, X.(
1999
). The meningococcal PilT protein is required for induction of intimate attachment to epithelial cells following pilus-mediated adhesion.
Proc. Natl. Acad. Sci. USA
96
,
4017
-4022.
Ring, A., Weiser, J. N. and Tuomanen, E. I.(
1998
). Pneumococcal trafficking across the blood-brain barrier. Molecular analysis of a novel bidirectional pathway.
J. Clinical Invest.
102
,
347
-360.
Rudel, T., Scheuerpflug, I. and Meyer, T. F.(
1995
). Neisseria PilC protein identified as type-4 pilus tip-located adhesin.
Nature
373
,
357
-362.
Schweitzer, K. M., Vicart, P., Delouis, C., Paulin, D., Drager,A. M., Langenhuijsen, M. M. and Weksler, B. B. (
2000
). Dynamics and segregation of cell-matrix adhesions in cultured fibroblasts.
Nat. Cell Biol.
2
,
191
-196.
Skoudy, A., Nhieu, G. T., Mantis, N., Arpin, M., Mounier, J.,Gounon, P. and Sansonetti, P. (
1999
). A functional role for ezrin during Shigella flexneri entry into epithelial cells.
J. Cell Sci.
112
,
2059
-2068.
Song, W. X., Ma, L., Chen, R. W. and Stein, D. C.(
2000
). Role of lipooligosaccharide in Opa-independent invasion of Neisseria gonorrhoeae into human epithelial cells.
J. Exp. Medicine
191
,
949
-959.
Virji, M., Makepeace, K., Ferguson, D. J. P., Achtman, M.,Sarkari, J. and Moxon, E. R. (
1992
). Expression of the Opc protein correlates with invasion of epithelial and endothelial cells by Neisseria meningitidis.
Mol. Microbiol
.
6
,
2785
-2795.
Virji, M., Makepeace, K., Ferguson, D. J. P., Achtman, M. and Moxon, E. R. (
1993
). Meningococcal Opa and Opc proteins:their role in colonization and invasion of human epithelial and endothelial cells.
Mol. Microbiol.
10
,
499
-510.
Virji, M., Watt, S. M., Barker, S., Makepeace, K. and Doyonnas,R. (
1996
). The N-domain of the humain CD66a adhesion molecule is a target for Opa proteins of Neisseria meningitidis and Neisseria gonorrhoeae.
Mol. Microb
.
22
,
929
-939.
Yamada, K. M. and Geiger, B. (
1997
). Molecular interactions in cell adhesion complexes.
Curr. Opin. Cell Biol.
9
,
76
-85.
Yonemura, S. and Tsukita, S. (
1999
). Direct involvement of ezrin/radixin/moesin (ERM)-binding membrane proteins in the organization of microvilli in collaboration with activated ERM proteins.
J. Cell Biol.
145
,
1497
-1509.
Yonemura, S., Hirao, M., Doi, Y., Takahashi, N., Kondo, T. and Tsukita, S. (
1998
). Ezrin/radixin/moesin (ERM) proteins bind to a positively charged amino acid cluster in the juxta-membrane cytoplasmic domain of CD44, CD43, and ICAM-2,
J. Cell Biol.
140
,
885
-895.
Zamir, E., Katz, B. Z., Aota, S., Yamada, K. M., Geiger, B. and Kam, Z. (
1999
). Molecular diversity of cell-matrix adhesions.
J. Cell Sci.
112
,
1655
-1669.