Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Infect Immun ; 83(1): 354-63, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25368118

ABSTRACT

Neisseria meningitidis asymptomatically colonizes the human upper respiratory tract but is also the cause of meningitis and severe septicemia. Carriage or disease evokes an immune response against the infecting strain. Hitherto, we have known little about the breadth of immunity induced by natural carriage of a single strain or its implications for subsequent infectious challenge. In this study, we establish that transgenic mice expressing human CEACAM1 support nasal colonization by a variety of strains of different capsular types. Next, we nasally challenged these mice with either of the N. meningitidis strains H44/76 (serogroup B, ST-32) and 90/18311 (serogroup C, ST-11), while following the induction of strain-specific immunoglobulin. When these antisera were tested for reactivity with a diverse panel of N. meningitidis strains, very low levels of antibody were detected against all meningococcal strains, yet a mutually exclusive "fingerprint" of high-level cross-reactivity toward certain strains became apparent. To test the efficacy of these responses for protection against subsequent challenge, CEACAM1-humanized mice exposed to strain 90/18311 were then rechallenged with different N. meningitidis strains. As expected, the mice were immune to challenge with the same strain and with a closely related ST-11 strain, 38VI, while H44/76 (ST-32) could still colonize these animals. Notably, however, despite the paucity of detectable humoral response against strain 196/87 (ST-32), this strain was unable to colonize the 90/18311-exposed mice. Combined, our data suggest that current approaches may underestimate the actual breadth of mucosal protection gained through natural exposure to N. meningitidis strains.


Subject(s)
Antibodies, Bacterial/blood , Antigens, CD/immunology , Carrier State/immunology , Cell Adhesion Molecules/immunology , Meningococcal Infections/immunology , Meningococcal Infections/prevention & control , Neisseria meningitidis/immunology , Animals , Antigens, CD/genetics , Cell Adhesion Molecules/genetics , Humans , Mice, Transgenic
2.
J Nat Prod ; 77(10): 2170-81, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25255326

ABSTRACT

The first total synthesis of all six known A54556 acyldepsipeptide (ADEP) antibiotics from Streptomyces hawaiiensis is reported. This family of compounds has a unique mechanism of antibacterial action, acting as activators of caseinolytic protease (ClpP). Assembly of the 16-membered depsipeptide core was accomplished via a pentafluorophenyl ester-based macrolactamization strategy. Late stage amine deprotection was carried out under neutral conditions by employing a mild hydrogenolysis strategy, which avoids the formation of undesired ring-opened depsipeptide side products encountered during deprotection of acid-labile protecting groups. The free amines were found to be significantly more reactive toward late stage amide bond formation as compared to the corresponding ammonium salts, giving final products in excellent yields. A thorough NMR spectroscopic analysis of these compounds was carried out to formally assign the structures and to aid with the spectroscopic assignment of ADEP analogues. The identity of two of the structures was confirmed by comparison with biologically produced samples from S. hawaiiensis. An X-ray crystallographic analysis of an ADEP analogue reveals a conformation similar to that found in cocrystal structures of ADEPs with ClpP protease. The degree of antibacterial activity of the different compounds was evaluated in vitro using MIC assays employing both Gram-positive and Gram-negative strains and a fluorescence-based biochemical assay.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Depsipeptides/chemical synthesis , Depsipeptides/pharmacology , Streptomyces/chemistry , Anti-Bacterial Agents/chemistry , Crystallography, X-Ray , Depsipeptides/chemistry , Endopeptidase Clp , Escherichia coli Proteins/agonists , Microbial Sensitivity Tests , Molecular Structure , Neisseria meningitidis/drug effects , Nuclear Magnetic Resonance, Biomolecular , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects
3.
PLoS Pathog ; 9(7): e1003509, 2013.
Article in English | MEDLINE | ID: mdl-23935487

ABSTRACT

Neisseria meningitidis (Nme) asymptomatically colonizes the human nasopharynx, yet can initiate rapidly-progressing sepsis and meningitis in rare instances. Understanding the meningococcal lifestyle within the nasopharyngeal mucosa, a phase of infection that is prerequisite for disease, has been hampered by the lack of animal models. Herein, we compare mice expressing the four different human carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) that can bind the neisserial Opa protein adhesins, and find that expression of human CEACAM1 is necessary and sufficient to establish intranasal colonization. During infection, in vivo selection for phase variants expressing CEACAM1-specific Opa proteins occurs, allowing mucosal attachment and entry into the subepithelial space. Consistent with an essential role for Opa proteins in this process, Opa-deficient meningococci were unable to colonize the CEACAM1-humanized mice. While simple Opa-mediated attachment triggered an innate response regardless of meningococcal viability within the inoculum, persistence of viable Opa-expressing bacteria within the CEACAM1-humanized mice was required for a protective memory response to be achieved. Parenteral immunization with a capsule-based conjugate vaccine led to the accumulation of protective levels of Nme-specific IgG within the nasal mucus, yet the sterilizing immunity afforded by natural colonization was instead conferred by Nme-specific IgA without detectable IgG. Considered together, this study establishes that the availability of CEACAM1 helps define the exquisite host specificity of this human-restricted pathogen, displays a striking example of in vivo selection for the expression of desirable Opa variants, and provides a novel model in which to consider meningococcal infection and immunity within the nasopharyngeal mucosa.


Subject(s)
Adaptation, Physiological , Meningococcal Infections/microbiology , Nasopharynx/microbiology , Neisseria meningitidis/growth & development , Respiratory Mucosa/microbiology , Respiratory Tract Infections/microbiology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Bacterial Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Escherichia coli/metabolism , HeLa Cells , Humans , Immunity, Mucosal , Meningococcal Infections/immunology , Meningococcal Infections/metabolism , Mice , Mice, Transgenic , Microbial Viability , Mutation , Nasopharynx/immunology , Nasopharynx/metabolism , Nasopharynx/pathology , Neisseria meningitidis/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Neutrophils/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/pathology
4.
Proc Natl Acad Sci U S A ; 110(25): 10234-9, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23733950

ABSTRACT

Clinical and epidemiological synergy exists between the globally important sexually transmitted infections, gonorrhea and HIV. Neisseria gonorrhoeae, which causes gonorrhea, is particularly adept at driving HIV-1 expression, but the molecular determinants of this relationship remain undefined. N. gonorrhoeae liberates a soluble factor that potently induces expression from the HIV-1 LTR in coinfected cluster of differentiation 4-positive (CD4(+)) T lymphocytes, but this factor is not a previously described innate effector. A genome-wide mutagenesis approach was undertaken to reveal which component(s) of N. gonorrhoeae induce HIV-1 expression in CD4(+) T lymphocytes. A mutation in the ADP-heptose biosynthesis gene, hldA, rendered the bacteria unable to induce HIV-1 expression. The hldA mutant has a truncated lipooligosaccharide structure, contains lipid A in its outer membrane, and remains bioactive in a TLR4 reporter-based assay but did not induce HIV-1 expression. Mass spectrometry analysis of extensively fractionated N. gonorrhoeae-derived supernatants revealed that the LTR-inducing fraction contained a compound having a mass consistent with heptose-monophosphate (HMP). Heptose is a carbohydrate common in microbes but is absent from the mammalian glycome. Although ADP-heptose biosynthesis is common among Gram-negative bacteria, and heptose is a core component of most lipopolysaccharides, N. gonorrhoeae is peculiar in that it effectively liberates HMP during growth. This N. gonorrhoeae-derived HMP activates CD4(+) T cells to invoke an NF-κB-dependent transcriptional response that drives HIV-1 expression and viral production. Our study thereby shows that heptose is a microbial-specific product that is sensed as an innate immune agonist and unveils the molecular link between N. gonorrhoeae and HIV-1.


Subject(s)
Coinfection/immunology , Gonorrhea , HIV Infections , HIV-1/enzymology , Heptoses/immunology , Neisseria gonorrhoeae/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/virology , Female , Gonorrhea/immunology , Gonorrhea/microbiology , Gonorrhea/virology , HIV Infections/immunology , HIV Infections/microbiology , HIV Infections/virology , HIV Long Terminal Repeat/genetics , HIV-1/immunology , Heptoses/genetics , Heptoses/metabolism , Humans , Jurkat Cells , Male , Neisseria gonorrhoeae/immunology , Toll-Like Receptor 5/immunology
5.
PLoS One ; 8(2): e56705, 2013.
Article in English | MEDLINE | ID: mdl-23424672

ABSTRACT

Infection with Neisseria gonorrhoeae (N. gonorrhoeae) can trigger an intense local inflammatory response at the site of infection, yet there is little specific immune response or development of immune memory. Gonococcal surface epitopes are known to undergo antigenic variation; however, this is unlikely to explain the weak immune response to infection since individuals can be re-infected by the same serotype. Previous studies have demonstrated that the colony opacity-associated (Opa) proteins on the N. gonorrhoeae surface can bind human carcinoembryonic antigen-related cellular adhesion molecule 1 (CEACAM1) on CD4⁺ T cells to suppress T cell activation and proliferation. Interesting in this regard, N. gonorrhoeae infection is associated with impaired HIV-1 (human immunodeficiency virus type 1)-specific cytotoxic T-lymphocyte (CTL) responses and with transient increases in plasma viremia in HIV-1-infected patients, suggesting that N. gonorrhoeae may also subvert immune responses to co-pathogens. Since dendritic cells (DCs) are professional antigen presenting cells (APCs) that play a key role in the induction of an adaptive immune response, we investigated the effects of N. gonorrhoeae Opa proteins on human DC activation and function. While morphological changes reminiscent of DC maturation were evident upon N. gonorrhoeae infection, we observed a marked downregulation of DC maturation marker CD83 when the gonococci expressing CEACAM1-specific Opa(CEA), but not other Opa variants. Consistent with a gonococcal-induced defect in maturation, Opa(CEA) binding to CEACAM1 reduced the DCs' capacity to stimulate an allogeneic T cell proliferative response. Moreover, Opa(CEA)-expressing N. gonorrhoeae showed the potential to impair DC-dependent development of specific adaptive immunity, since infection with Opa(CEA)-positive gonococci suppressed the ability of DCs to stimulate HIV-1-specific memory CTL responses. These results reveal a novel mechanism to explain why infection of N. gonorrhoeae fails to trigger an effective specific immune response or develop immune memory, and may affect the potent synergy between gonorrhea and HIV-1 infection.


Subject(s)
Antigens, CD/metabolism , Bacterial Outer Membrane Proteins/metabolism , Cell Adhesion Molecules/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , HIV-1/immunology , Neisseria gonorrhoeae/metabolism , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity , Cell Proliferation , Dendritic Cells/microbiology , Dendritic Cells/virology , Down-Regulation , Fimbriae, Bacterial/physiology , Humans , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Monocytes/cytology , Neisseria gonorrhoeae/physiology , Species Specificity , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/virology , CD83 Antigen
6.
Infect Immun ; 80(7): 2346-53, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22508859

ABSTRACT

The capsule of Neisseria meningitidis is the major virulence factor that enables this bacterium to overcome host immunity elicited by complement and phagocytes, rendering it capable of surviving in blood. As such, nonencapsulated N. meningitidis isolates are generally considered nonpathogenic. Here, we consider the inherent virulence of two nonencapsulated N. meningitidis isolates obtained from our national surveillance of infected blood cultures in Canada. Capsule deficiency of both strains was confirmed by serology and PCR for the ctrA to ctrD genes and siaA to siaC genes, as well as siaD genes specific to serogroups B, C, Y, and W135. In both strains, the capsule synthesis genes were replaced by the capsule null locus, cnl-2. In accordance with a lack of capsule, both strains were fully susceptible to killing by both human and baby rabbit complement. However, in the presence of cytidine-5' monophospho-N-acetylneuraminic acid (CMP-NANA), allowing for lipooligosaccharide (LOS) sialylation, a significant increase of resistance to complement killing was observed. Mass spectrometry of purified LOS did not reveal any uncommon modifications that would explain their invasive phenotype. Finally, in a mouse intraperitoneal challenge model, these nonencapsulated isolates displayed enhanced virulence relative to an isogenic mutant of serogroup B strain MC58 lacking capsule (MC58ΔsiaD). Virulence of all nonencapsulated isolates tested was below that of encapsulated serogroup B strains MC58 and B16B6. However, whereas no mortality was observed with MC58ΔsiaD, 5/10 mice succumbed to infection with strain 2275 and 2/11 mice succumbed to strain 2274. Our results suggest the acquisition of a new virulence phenotype by these nonencapsulated strains.


Subject(s)
Bacterial Capsules/genetics , Bacterial Capsules/metabolism , Meningococcal Infections/microbiology , Neisseria meningitidis/pathogenicity , Virulence Factors/deficiency , Virulence Factors/metabolism , Animals , Bacterial Capsules/immunology , Canada , Complement System Proteins/immunology , Disease Models, Animal , Female , Genes, Bacterial , Humans , Male , Meningococcal Infections/mortality , Mice , Mice, Inbred C57BL , Neisseria meningitidis/immunology , Neisseria meningitidis/isolation & purification , Peritonitis/microbiology , Peritonitis/mortality , Polymerase Chain Reaction , Rabbits , Serotyping , Survival Analysis , Virulence Factors/immunology
7.
Chem Biol ; 18(9): 1167-78, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21944755

ABSTRACT

ClpP is a cylindrical serine protease whose ability to degrade proteins is regulated by the unfoldase ATP-dependent chaperones. ClpP on its own can only degrade small peptides. Here, we used ClpP as a target in a high-throughput screen for compounds, which activate the protease and allow it to degrade larger proteins, hence, abolishing the specificity arising from the ATP-dependent chaperones. Our screen resulted in five distinct compounds, which we designate as Activators of Self-Compartmentalizing Proteases 1 to 5 (ACP1 to 5). The compounds are found to stabilize the ClpP double-ring structure. The ACP1 chemical structure was considered to have drug-like characteristics and was further optimized to give analogs with bactericidal activity. Hence, the ACPs represent classes of compounds that can activate ClpP and that can be developed as potential novel antibiotics.


Subject(s)
Anti-Bacterial Agents/chemistry , Endopeptidase Clp/chemistry , Escherichia coli Proteins/chemistry , Anti-Bacterial Agents/pharmacology , Binding Sites , Computer Simulation , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Enzyme Activation/drug effects , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Microbial Sensitivity Tests , Molecular Chaperones/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
8.
Cell Microbiol ; 9(9): 2153-66, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17506821

ABSTRACT

Mature, microbicidal phagosomes are rich in the lysosome-associated membrane proteins, LAMP-1 and LAMP-2, two highly glycosylated proteins presumed to form a protective barrier lining the phagosomal membrane. Pathogenic Neisseria secrete a protease that selectively cleaves LAMP-1, suggesting a critical role for LAMP proteins in the microbicidal competence of phagosomes. To determine the requirement for LAMP proteins in bacterial phagocytosis, we employed embryonic fibroblasts isolated from knockout mice lacking lamp-1, lamp-2 or both genes, as well as small interfering RNA (siRNA)-mediated knockdown of LAMP expression in a human epithelial cell line. Like wild-type cells, those lacking either LAMP-1 or LAMP-2 alone formed phagosomes that gradually acquired microbicidal activity and curtailed bacterial growth. In contrast, LAMP-1 and LAMP-2 double-deficient fibroblasts failed to kill engulfed Neisseria gonorrhoeae. In these cells, maturation was arrested prior to the acquisition of Rab7. As a result, the Rab7-interacting lysosomal protein (RILP, a Rab7 effector) was not recruited to the phagosomes, which were consequently unable to undergo dynein/dynactin-mediated centripetal displacement along microtubules and remained in a predominantly peripheral location. The inability of such phagosomes to migrate towards lysosomes likely contributed to their incomplete maturation and inability to eliminate bacteria. These findings suggest that neisserial degradation of LAMP-1 is not sufficient to affect its survival within the phagosome, and establish LAMP proteins as critical components in the process whereby phagosomes acquire microbicidal capabilities.


Subject(s)
Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Neisseria gonorrhoeae , Phagosomes , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/metabolism , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/genetics , Mice , Mice, Knockout , Neisseria gonorrhoeae/metabolism , Neisseria gonorrhoeae/pathogenicity , Phagocytosis/physiology , Phagosomes/metabolism , Phagosomes/microbiology , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tetraspanin 30 , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
9.
Infect Immun ; 72(5): 2742-52, 2004 May.
Article in English | MEDLINE | ID: mdl-15102784

ABSTRACT

Individual Neisseria gonorrhoeae colony opacity-associated (Opa) protein variants can bind up to four different carcinoembryonic antigen-related cellular adhesion molecule (CEACAM) receptors. Most human cells encountered by gonococci express a combination of CEACAM receptors, thereby complicating the elucidation of intracellular signaling pathways triggered by individual receptors. Here, we compare the process of bacterial engulfment by a panel of stably transfected HeLa epithelial cell lines expressing each CEACAM receptor in isolation. CEACAM1 and CEACAM3 each contain proteinaceous transmembrane and cytoplasmic domains; however, the processes of neisserial uptake mediated by these receptors differ with respect to their susceptibilities to both tyrosine kinase inhibitors and the actin microfilament-disrupting agent cytochalasin D. Neisserial uptake mediated by glycosylphosphatidylinositol (GPI)-anchored CEACAM5 and CEACAM6 was not significantly affected by any of a broad spectrum of inhibitors tested. However, cleavage of the GPI anchor by phosphatidylinositol-specific phospholipase C reduced bacterial uptake by HeLa cells expressing CEACAM5, consistent with a single zipper-like mechanism of uptake mediated by this receptor. Regardless of the CEACAM receptor expressed, internalized gonococci were effectively killed by a microtubule-dependent process that required acidification of the bacterium-containing phagosome. Given the phase-variable nature of neisserial Opa proteins, these results indicate that the mechanism of bacterial engulfment and the cellular response to gonococcal infection depend on both the receptor specificities of the neisserial Opa protein variants expressed and the spectrum of CEACAM receptors present on target cells, each of which determines the combination of receptors ultimately engaged.


Subject(s)
Bacterial Adhesion/immunology , Integrins/physiology , Neisseria gonorrhoeae/immunology , Antigens, CD/genetics , Antigens, CD/physiology , Antigens, Differentiation/genetics , Antigens, Differentiation/physiology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/physiology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/physiology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cytoskeleton/physiology , GPI-Linked Proteins , Glycosylphosphatidylinositols/physiology , HeLa Cells , Humans , Integrins/genetics , Kinetics , Models, Biological , Neisseria gonorrhoeae/pathogenicity , Phagocytosis , Phosphorylation , Transfection
10.
Mol Microbiol ; 49(3): 623-37, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12864848

ABSTRACT

Gonorrhea is characterized by a purulent urethral or cervical discharge consisting primarily of neutrophils associated with Neisseria gonorrhoeae. These interactions are facilitated by gonococcal colony opacity-associated (Opa) protein binding to host cellular CEACAM receptors. Of these, CEACAM3 is restricted to neutrophils and contains an immunoreceptor tyrosine-based activation motif (ITAM) reminiscent of that found within certain phagocytic Fc receptors. CEACAM3 was tyrosine phosphorylated by a Src family kinase-dependent process upon infection by gonococci expressing CEACAM-specific Opa proteins. This phosphorylation was necessary for efficient bacterial uptake; however, a less efficient uptake process became evident when kinase inhibitors or mutagenesis of the ITAM were used to prevent phosphorylation. Ligated CEACAM3 was recruited to a cytoskeleton-containing fraction, intense foci of polymerized actin were evident where bacteria attached to HeLa-CEACAM3, and disruption of polymerized actin by cytochalasin D blocked all bacterial uptake by these cells. These data support a model whereby CEACAM3 can mediate the Opa-dependent uptake of N. gonorrhoeae via either an efficient, ITAM phosphorylation-dependent process that resembles phagocytosis or a less efficient, tyrosine phosphorylation-independent mechanism.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Carcinoembryonic Antigen/metabolism , Neisseria gonorrhoeae/metabolism , Neutrophils/metabolism , Protein Processing, Post-Translational , Proteoglycans/metabolism , Receptors, Cell Surface/metabolism , src-Family Kinases/metabolism , Actin Cytoskeleton/ultrastructure , Adhesins, Bacterial/genetics , Amino Acid Motifs , Amino Acid Sequence , Antigens, Bacterial , Bacterial Proteins/genetics , Carcinoembryonic Antigen/chemistry , Carcinoembryonic Antigen/genetics , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , HeLa Cells/metabolism , HeLa Cells/microbiology , Humans , Molecular Sequence Data , Neisseria gonorrhoeae/genetics , Neutrophils/microbiology , Neutrophils/ultrastructure , Phosphorylation , Proteoglycans/genetics , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/metabolism , Transfection
11.
J Biol Chem ; 278(16): 14037-45, 2003 Apr 18.
Article in English | MEDLINE | ID: mdl-12571236

ABSTRACT

Neisseria gonorrhoeae can be internalized by mammalian cells through interactions between bacterial opacity-associated (Opa) adhesins and members of the human carcinoembryonic antigen-related cellular adhesion molecule (CEACAM) family. We examined the role of phosphatidylinositol 3-kinases (PI3Ks) in gonococcal invasion of epithelial cell lines expressing either CEACAM1 or CEACAM3. CEACAM3-mediated internalization, but not that mediated by CEACAM1, was accompanied by localized and transient accumulation of the class I PI3K product phosphatidylinositol 3,4,5-trisphosphate at sites of bacterial engulfment. Inhibition of phosphatidylinositol 3-kinases reduced CEACAM3-mediated uptake but, paradoxically, led to an increase in intracellular survival of bacteria internalized via either CEACAM1 or CEACAM3, suggesting additional roles for PI3K products. Consistent with this finding, the class III PI3K product phosphatidylinositol 3-phosphate accumulated and persisted in the membrane of gonococcal phagosomes after internalization. Inhibition of PI3K blocked phagosomal acquisition of the late endosomal marker lysosome-associated membrane protein 2 and reduced phagosomal acidification. Inhibiting phagosomal acidification with concanamycin A also increased survival of intracellular gonococci. These results suggest two modes of action of phosphatidylinositol 3-kinases during internalization of gonococci: synthesis of phosphatidylinositol 3,4,5-trisphosphate is important for CEACAM3-mediated uptake, while phosphatidylinositol 3-phosphate is needed for phagosomal maturation and acidification, which are required for optimal bacterial killing.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Carcinoembryonic Antigen/metabolism , Macrolides , Phosphatidylinositol 3-Kinases/physiology , Androstadienes/pharmacology , Anti-Bacterial Agents/pharmacology , Cell Adhesion Molecules , Cell Division , Cell Line , Cell Nucleus/metabolism , Chromones/pharmacology , Drug Resistance , Endosomes/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gentamicins/pharmacology , HeLa Cells , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Microscopy, Confocal , Morpholines/pharmacology , Neisseria gonorrhoeae/metabolism , Phagosomes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Plasmids/metabolism , Time Factors , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...