Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
mBio ; 11(6)2020 12 08.
Article in English | MEDLINE | ID: mdl-33293379

ABSTRACT

Subversion of heparan sulfate proteoglycans (HSPGs) is thought to be a common virulence mechanism shared by many microbial pathogens. The prevailing assumption is that pathogens co-opt HSPGs as cell surface attachment receptors or as inhibitors of innate host defense. However, there are few data that clearly support this idea in vivo We found that deletion of syndecan-1 (Sdc1), a major cell surface HSPG of epithelial cells, causes a gain of function in a mouse model of scarified corneal infection, where Sdc1-/- corneas were significantly less susceptible to Streptococcus pneumoniae infection. Administration of excess Sdc1 ectodomains significantly inhibited S. pneumoniae corneal infection, suggesting that Sdc1 promotes infection as a cell surface attachment receptor. However, S. pneumoniae did not interact with Sdc1 and Sdc1 was shed upon S. pneumoniae infection, indicating that Sdc1 does not directly support S. pneumoniae adhesion. Instead, Sdc1 promoted S. pneumoniae adhesion by driving the assembly of fibronectin (FN) fibrils in the corneal basement membrane to which S. pneumoniae attaches when infecting injured corneas. S. pneumoniae specifically bound to corneal FN via PavA, and PavA deletion significantly attenuated S. pneumoniae virulence in the cornea. Excess Sdc1 ectodomains inhibited S. pneumoniae corneal infection by binding to the Hep II domain and interfering with S. pneumoniae PavA binding to FN. These findings reveal a previously unknown virulence mechanism of S. pneumoniae where key extracellular matrix (ECM) interactions and structures that are essential for host cell homeostasis are exploited for bacterial pathogenesis.IMPORTANCE Bacterial pathogens have evolved several ingenious mechanisms to subvert host cell biology for their pathogenesis. Bacterial attachment to the host ECM establishes a niche to grow and is considered one of the critical steps of infection. This pathogenic mechanism entails coordinated assembly of the ECM by the host to form the ECM structure and organization that are specifically recognized by bacteria for their adhesion. We serendipitously discovered that epithelial Sdc1 facilitates the assembly of FN fibrils in the corneal basement membrane and that this normal biological function of Sdc1 has detrimental consequences for the host in S. pneumoniae corneal infection. Our studies suggest that bacterial subversion of the host ECM is more complex than previously appreciated.


Subject(s)
Fibronectins/metabolism , Host-Pathogen Interactions , Keratitis/metabolism , Keratitis/microbiology , Streptococcus pneumoniae/physiology , Syndecan-1/metabolism , Animals , Bacterial Adhesion , Cornea/metabolism , Cornea/microbiology , Cornea/pathology , Disease Models, Animal , Extracellular Matrix/metabolism , Fluorescent Antibody Technique , Gain of Function Mutation , Gene Expression , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Host-Pathogen Interactions/genetics , Immunohistochemistry , Keratitis/pathology , Mice , Mice, Knockout , Syndecan-1/genetics
2.
J Biol Chem ; 295(19): 6689-6699, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32229583

ABSTRACT

The cell surfaces of many bacteria carry filamentous polypeptides termed adhesins that enable binding to both biotic and abiotic surfaces. Surface adherence is facilitated by the exquisite selectivity of the adhesins for their cognate ligands or receptors and is a key step in niche or host colonization and pathogenicity. Streptococcus gordonii is a primary colonizer of the human oral cavity and an opportunistic pathogen, as well as a leading cause of infective endocarditis in humans. The fibrillar adhesin CshA is an important determinant of S. gordonii adherence, forming peritrichous fibrils on its surface that bind host cells and other microorganisms. CshA possesses a distinctive multidomain architecture comprising an N-terminal target-binding region fused to 17 repeat domains (RDs) that are each ∼100 amino acids long. Here, using structural and biophysical methods, we demonstrate that the intact CshA repeat region (CshA_RD1-17, domains 1-17) forms an extended polymeric monomer in solution. We recombinantly produced a subset of CshA RDs and found that they differ in stability and unfolding behavior. The NMR structure of CshA_RD13 revealed a hitherto unreported all ß-fold, flanked by disordered interdomain linkers. These findings, in tandem with complementary hydrodynamic studies of CshA_RD1-17, indicate that this polypeptide possesses a highly unusual dynamic transitory structure characterized by alternating regions of order and disorder. This architecture provides flexibility for the adhesive tip of the CshA fibril to maintain bacterial attachment that withstands shear forces within the human host. It may also help mitigate deleterious folding events between neighboring RDs that share significant structural identity without compromising mechanical stability.


Subject(s)
Bacterial Proteins/chemistry , Membrane Proteins/chemistry , Protein Multimerization , Amino Acid Sequence , Models, Molecular , Protein Domains , Protein Structure, Quaternary , Repetitive Sequences, Amino Acid
3.
PLoS Pathog ; 15(6): e1007848, 2019 06.
Article in English | MEDLINE | ID: mdl-31181121

ABSTRACT

Streptococcus agalactiae (Group B Streptococcus, GBS) normally colonizes healthy adults but can cause invasive disease, such as meningitis, in the newborn. To gain access to the central nervous system, GBS must interact with and penetrate brain or meningeal blood vessels; however, the exact mechanisms are still being elucidated. Here, we investigate the contribution of BspC, an antigen I/II family adhesin, to the pathogenesis of GBS meningitis. Disruption of the bspC gene reduced GBS adherence to human cerebral microvascular endothelial cells (hCMEC), while heterologous expression of BspC in non-adherent Lactococcus lactis conferred bacterial attachment. In a murine model of hematogenous meningitis, mice infected with ΔbspC mutants exhibited lower mortality as well as decreased brain bacterial counts and inflammatory infiltrate compared to mice infected with WT GBS strains. Further, BspC was both necessary and sufficient to induce neutrophil chemokine expression. We determined that BspC interacts with the host cytoskeleton component vimentin and confirmed this interaction using a bacterial two-hybrid assay, microscale thermophoresis, immunofluorescent staining, and imaging flow cytometry. Vimentin null mice were protected from WT GBS infection and also exhibited less inflammatory cytokine production in brain tissue. These results suggest that BspC and the vimentin interaction is critical for the pathogenesis of GBS meningitis.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Brain/metabolism , Meningitis, Bacterial/metabolism , Streptococcal Infections/metabolism , Streptococcus agalactiae/metabolism , Vimentin/metabolism , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Brain/blood supply , Brain/microbiology , Brain/pathology , Endothelium, Vascular , HeLa Cells , Humans , Male , Meningitis, Bacterial/genetics , Meningitis, Bacterial/pathology , Mice , Mice, Mutant Strains , Sheep , Streptococcal Infections/genetics , Streptococcal Infections/pathology , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Vimentin/genetics
4.
PLoS One ; 13(12): e0208317, 2018.
Article in English | MEDLINE | ID: mdl-30513116

ABSTRACT

Saliva contains many proteins that have an important role in biological process of the oral cavity and is closely associated with many diseases. Although the dog is a common companion animal, the composition of salivary proteome and its relationship with that of human are unclear. In this study, shotgun proteomics was used to compare the salivary proteomes of 7 Thai village dogs and 7 human subjects. Salivary proteomes revealed 2,532 differentially expressed proteins in dogs and humans, representing various functions including cellular component organization or biogenesis, cellular process, localization, biological regulation, response to stimulus, developmental process, multicellular organismal process, metabolic process, immune system process, apoptosis and biological adhesion. The oral proteomes of dogs and humans were appreciably different. Proteins related to apoptosis processes and biological adhesion were predominated in dog saliva. Drug-target network predictions by STITCH Version 5.0 showed that dog salivary proteins were found to have potential roles in tumorigenesis, anti-inflammation and antimicrobial processes. In addition, proteins related to regeneration and healing processes such as fibroblast growth factor and epidermal growth factor were also up-regulated in dogs. These findings provide new information on dog saliva composition and will be beneficial for the study of dog saliva in diseased and health conditions in the future.


Subject(s)
Proteomics/methods , Saliva/metabolism , Salivary Proteins and Peptides/metabolism , Amino Acid Sequence , Animals , Dogs , Humans , Salivary Proteins and Peptides/chemistry
6.
mBio ; 9(2)2018 04 24.
Article in English | MEDLINE | ID: mdl-29691333

ABSTRACT

The pleiomorphic yeast Candida albicans is a significant pathogen in immunocompromised individuals. In the oral cavity, C. albicans is an inhabitant of polymicrobial communities, and interspecies interactions promote hyphal formation and biofilm formation. C. albicans colonizes the subgingival area, and the frequency of colonization increases in periodontal disease. In this study, we investigated the interactions between C. albicans and the periodontal pathogen Porphyromonas gingivalisC. albicans and P. gingivalis were found to coadhere in both the planktonic and sessile phases. Loss of the internalin-family protein InlJ abrogated adhesion of P. gingivalis to C. albicans, and recombinant InlJ protein competitively inhibited interspecies binding. A mutant of C. albicans deficient in expression of major hyphal protein Als3 showed diminished binding to P. gingivalis, and InlJ interacted with Als3 heterologously expressed in Saccharomyces cerevisiae Transcriptional profiling by RNA sequencing (RNA-Seq) established that 57 genes were uniquely upregulated in an InlJ-dependent manner in P. gingivalis-C. albicans communities, with overrepresentation of those corresponding to 31 gene ontology terms, including those associated with growth and division. Of potential relevance to the disease process, C. albicans induced upregulation of components of the type IX secretion apparatus. Collectively, these findings indicate that InlJ-Als3-dependent binding facilitates interdomain community development between C. albicans and P. gingivalis and that P. gingivalis has the potential for increased virulence within such communities.IMPORTANCE Many diseases involve the concerted actions of microorganisms assembled in polymicrobial communities. Inflammatory periodontal diseases are among the most common infections of humans and result in destruction of gum tissue and, ultimately, in loss of teeth. In periodontal disease, pathogenic communities can include the fungus Candida albicans; however, the contribution of C. albicans to the synergistic virulence of the community is poorly understood. Here we characterize the interactions between C. albicans and the keystone bacterial pathogen Porphyromonas gingivalis and show that coadhesion mediated by specific proteins results in major changes in gene expression by P. gingivalis, which could serve to increase pathogenic potential. The work provides significant insights into interdomain interactions that can enhance our understanding of diseases involving a multiplicity of microbial pathogens.


Subject(s)
Bacterial Proteins/metabolism , Candida albicans/physiology , Fungal Proteins/metabolism , Microbial Interactions , Porphyromonas gingivalis/physiology , Biofilms/growth & development , Cell Adhesion , Gene Expression Profiling , Humans , Protein Binding
7.
Infect Immun ; 86(4)2018 04.
Article in English | MEDLINE | ID: mdl-29339458

ABSTRACT

Group B Streptococcus (GBS) is a leading cause of neonatal sepsis, pneumonia, and meningitis worldwide. In the majority of cases, GBS is transmitted vertically from mother to neonate, making maternal vaginal colonization a key risk factor for neonatal disease. The fungus Candida albicans is an opportunistic pathogen of the female genitourinary tract and the causative agent of vaginal thrush. Carriage of C. albicans has been shown to be an independent risk factor for vaginal colonization by GBS. However, the nature of interactions between these two microbes is poorly understood. This study provides evidence of a reciprocal, synergistic interplay between GBS and C. albicans that may serve to promote their cocolonization of the vaginal mucosa. GBS strains NEM316 (serotype III) and 515 (serotype Ia) are shown to physically interact with C. albicans, with the bacteria exhibiting tropism for candidal hyphal filaments. This interaction enhances association levels of both microbes with the vaginal epithelial cell line VK2/E6E7. The ability of GBS to coassociate with C. albicans is dependent upon expression of the hypha-specific adhesin Als3. In turn, expression of GBS antigen I/II family adhesins (Bsp polypeptides) facilitates this coassociation and confers upon surrogate Lactococcus lactis the capacity to exhibit enhanced interactions with C. albicans on vaginal epithelium. As genitourinary tract colonization is an essential first step in the pathogenesis of GBS and C. albicans, the coassociation mechanism reported here may have important implications for the risk of disease involving both of these pathogens.


Subject(s)
Candida albicans/immunology , Microbial Interactions , Mucous Membrane/immunology , Mucous Membrane/microbiology , Streptococcus agalactiae/immunology , Vagina/immunology , Vagina/microbiology , Adhesins, Bacterial/metabolism , Candida albicans/classification , Candida albicans/genetics , Candidiasis/immunology , Candidiasis/microbiology , Coinfection/immunology , Coinfection/microbiology , Epithelial Cells/microbiology , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , Mutation , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus agalactiae/classification , Streptococcus agalactiae/genetics
8.
Mol Microbiol ; 105(6): 839-859, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28657670

ABSTRACT

Colonization of mucosal respiratory surfaces is a prerequisite for the human pathobiont Streptococcus pneumoniae (the pneumococcus) to cause severe invasive infections. The arsenal of pneumococcal adhesins interacts with a multitude of extracellular matrix proteins. A paradigm for pneumococci is their interaction with the adhesive glycoprotein fibronectin, which facilitates bacterial adherence to host cells. Here, we deciphered the molecular interaction between fibronectin and pneumococcal fibronectin-binding proteins (FnBPs) PavA and PavB respectively. We show in adherence and binding studies that the pneumococcal interaction with fibronectin is a non-human specific trait. PavA and PavB target at least 13 out of 15 type III fibronectin domains as demonstrated in ligand overlay assays, surface plasmon resonance studies and SPOT peptide arrays. Strikingly, both pneumococcal FnBPs recognize similar peptides in targeted type III repeats. Structural comparisons revealed that the targeted type III repeat epitopes cluster on the inner strands of both ß-sheets forming the fibronectin domains. Importantly, synthetic peptides of FnIII1 , FnIII5 or FnIII15 bind directly to FnBPs PavA and PavB respectively. In conclusion, our study suggests a common pattern of molecular interactions between pneumococcal FnBPs and fibronectin. The specific epitopes recognized in this study can potentially be tested as antimicrobial targets in further scientific endeavours.


Subject(s)
Bacterial Proteins/metabolism , Fibronectin Type III Domain/physiology , Fibronectins/metabolism , Adhesins, Bacterial/metabolism , Bacterial Adhesion/genetics , Bacterial Adhesion/physiology , Bacterial Proteins/genetics , Carrier Proteins/metabolism , Fibronectin Type III Domain/genetics , Host-Pathogen Interactions , Humans , Protein Binding/genetics , Protein Binding/physiology , Protein Domains , Streptococcus pneumoniae/metabolism , Virulence Factors/metabolism
9.
Microbes Infect ; 19(7-8): 402-412, 2017.
Article in English | MEDLINE | ID: mdl-28456649

ABSTRACT

In Streptococcus pneumoniae TIGR4, genes encoding a SecY2A2 accessory Sec system are present within a locus encoding a serine-rich repeat surface protein PsrP. Mutant strains deleted in secA2 or psrP were deficient in biofilm formation, while the ΔsecA2 mutant was reduced in binding to airway epithelial cells. Cell wall protein (CWP) fractions from the ΔsecA2 mutant, but not from the ΔpsrP mutant, were reduced in haemolytic (pneumolysin) activity. Contact-dependent pneumolysin (Ply) activity of wild type TIGR4 cells was ten-fold greater than that of ΔsecA2 mutant cells suggesting that Ply was not active at the ΔsecA2 cell surface. Ply protein was found to be present in the CWP fraction from the ΔsecA2 mutant, but showed aberrant electrophoretic migration indicative of protein modification. Proteomic analyses led to the discovery that the ΔsecA2 mutant CWP fraction was deficient in two glycosidases as well as other enzymes involved in carbohydrate metabolism. Taken collectively the results suggest that positioning of Ply into the cell wall compartment in active form, together with glycosyl hydrolases and adhesins, requires a functional accessory Sec system.


Subject(s)
Bacterial Adhesion , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/metabolism , Biofilms/growth & development , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Streptolysins/metabolism , Bacterial Proteins/metabolism , Gene Deletion , Humans , Streptococcus pneumoniae/physiology
10.
J Clin Microbiol ; 55(6): 1837-1846, 2017 06.
Article in English | MEDLINE | ID: mdl-28356413

ABSTRACT

An emm32.2 invasive group A streptococcus (iGAS) outbreak occurred in Liverpool from January 2010 to September 2012. This genotype had not previously been identified in Liverpool, but was responsible for 32% (14/44) of all iGAS cases reported during this time period. We performed a case-case comparison of emm32.2 iGAS cases with non-emm32.2 control iGAS cases identified in the Liverpool population over the same time period to assess patient risk factors for emm32.2 iGAS infection. The emm32.2 iGAS cases were confined to the adult population. We show that homelessness, intravenous drug use, and alcohol abuse predisposed patients to emm32.2 iGAS disease; however, no obvious epidemiological linkage between the patients with emm32.2 iGAS could be identified. Comparative whole-genome sequencing analysis of emm32.2 iGAS and non-emm32.2 control isolates was also performed to identify pathogen factors which might have driven the outbreak. We identified 19 genes, five of which had previously been implicated in virulence, which were present in all of the emm32.2 iGAS isolates but not present in any of the non-emm32.2 control isolates. We report that a novel emm32.2 genotype emerged in Liverpool in 2010 and identified a specific subset of genes, which could have allowed this novel emm32.2 genotype to persist in a disadvantaged population in the region over a 3-year period.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/genetics , Disease Outbreaks , Genotype , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/classification , Streptococcus pyogenes/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Molecular Epidemiology , Streptococcus pyogenes/isolation & purification , United Kingdom/epidemiology , Whole Genome Sequencing , Young Adult
11.
Cell Microbiol ; 19(1)2017 01.
Article in English | MEDLINE | ID: mdl-27616700

ABSTRACT

A range of Streptococcus bacteria are able to interact with blood platelets to form a thrombus (clot). Streptococcus gordonii is ubiquitous within the human oral cavity and amongst the common pathogens isolated from subjects with infective endocarditis. Two cell surface proteins, Hsa and Platelet adherence protein A (PadA), in S. gordonii mediate adherence and activation of platelets. In this study, we demonstrate that PadA binds activated platelets and that an NGR (Asparagine-Glycine-Arginine) motif within a 657 amino acid residue N-terminal fragment of PadA is responsible for this, together with two other integrin-like recognition motifs RGT and AGD. PadA also acts in concert with Hsa to mediate binding of S. gordonii to cellular fibronectin and vitronectin, and to promote formation of biofilms. Evidence is presented that PadA and Hsa are each reliant on the other's active presentation on the bacterial cell surface, suggesting cooperativity in functions impacting both colonization and pathogenesis.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Extracellular Matrix/metabolism , Host-Pathogen Interactions , Platelet Activation , Streptococcus gordonii/pathogenicity , Virulence Factors/metabolism , Bacterial Adhesion , Biofilms/growth & development , Hemagglutinins, Viral , Humans , Membrane Proteins/metabolism , Streptococcus gordonii/growth & development , Streptococcus gordonii/physiology
12.
J Biol Chem ; 292(5): 1538-1549, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27920201

ABSTRACT

Adherence of bacteria to biotic or abiotic surfaces is a prerequisite for host colonization and represents an important step in microbial pathogenicity. This attachment is facilitated by bacterial adhesins at the cell surface. Because of their size and often elaborate multidomain architectures, these polypeptides represent challenging targets for detailed structural and functional characterization. The multifunctional fibrillar adhesin CshA, which mediates binding to both host molecules and other microorganisms, is an important determinant of colonization by Streptococcus gordonii, an oral commensal and opportunistic pathogen of animals and humans. CshA binds the high-molecular-weight glycoprotein fibronectin (Fn) via an N-terminal non-repetitive region, and this protein-protein interaction has been proposed to promote S. gordonii colonization at multiple sites within the host. However, the molecular details of how these two proteins interact have yet to be established. Here we present a structural description of the Fn binding N-terminal region of CshA, derived from a combination of X-ray crystallography, small angle X-ray scattering, and complementary biophysical methods. In vitro binding studies support a previously unreported two-state "catch-clamp" mechanism of Fn binding by CshA, in which the disordered N-terminal domain of CshA acts to "catch" Fn, via formation of a rapidly assembled but also readily dissociable pre-complex, enabling its neighboring ligand binding domain to tightly clamp the two polypeptides together. This study presents a new paradigm for target binding by a bacterial adhesin, the identification of which will inform future efforts toward the development of anti-adhesive agents that target S. gordonii and related streptococci.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Fibronectins/metabolism , Membrane Proteins/metabolism , Streptococcus gordonii/metabolism , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Fibronectins/chemistry , Fibronectins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Binding , Protein Domains , Streptococcus gordonii/chemistry , Streptococcus gordonii/genetics
13.
J Biol Chem ; 291(41): 21474-21484, 2016 Oct 07.
Article in English | MEDLINE | ID: mdl-27551046

ABSTRACT

The accessory Sec system in Streptococcus gordonii DL1 is a specialized export system that transports a large serine-rich repeat protein, Hsa, to the bacterial surface. The system is composed of core proteins SecA2 and SecY2 and accessory Sec proteins Asp1-Asp5. Similar to canonical SecYEG, SecY2 forms a channel for translocation of the Hsa adhesin across the cytoplasmic membrane. Accessory Sec proteins Asp4 and Asp5 have been suggested to work alongside SecY2 to form the translocon, similar to the associated SecY, SecE, and SecG of the canonical system (SecYEG). To test this theory, S. gordonii secY2, asp4, and asp5 were co-expressed in Escherichia coli The resultant complex was subsequently purified, and its composition was confirmed by mass spectrometry to be SecY2-Asp4-Asp5. Like SecYEG, the non-canonical complex activates the ATPase activity of the SecA motor (SecA2). This study also shows that Asp4 and Asp5 are necessary for optimal adhesion of S. gordonii to glycoproteins gp340 and fibronectin, known Hsa binding partners, as well as for early stage biofilm formation. This work opens new avenues for understanding the structure and function of the accessory Sec system.


Subject(s)
Bacterial Proteins , SEC Translocation Channels , Streptococcus gordonii , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium-Binding Proteins , DNA-Binding Proteins , Humans , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , SEC Translocation Channels/chemistry , SEC Translocation Channels/genetics , SEC Translocation Channels/metabolism , Streptococcus gordonii/chemistry , Streptococcus gordonii/genetics , Streptococcus gordonii/metabolism , Tumor Suppressor Proteins
14.
J Biol Chem ; 291(31): 15985-6000, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27311712

ABSTRACT

Streptococcus agalactiae (group B Streptococcus, GBS) is the predominant cause of early-onset infectious disease in neonates and is responsible for life-threatening infections in elderly and immunocompromised individuals. Clinical manifestations of GBS infection include sepsis, pneumonia, and meningitis. Here, we describe BspA, a deviant antigen I/II family polypeptide that confers adhesive properties linked to pathogenesis in GBS. Heterologous expression of BspA on the surface of the non-adherent bacterium Lactococcus lactis confers adherence to scavenger receptor gp340, human vaginal epithelium, and to the fungus Candida albicans Complementary crystallographic and biophysical characterization of BspA reveal a novel ß-sandwich adhesion domain and unique asparagine-dependent super-helical stalk. Collectively, these findings establish a new bacterial adhesin structure that has in effect been hijacked by a pathogenic Streptococcus species to provide competitive advantage in human mucosal infections.


Subject(s)
Adhesins, Bacterial/chemistry , Bacterial Proteins/chemistry , Cell Wall/chemistry , Membrane Proteins/chemistry , Streptococcus agalactiae/chemistry , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Candida albicans/genetics , Candida albicans/metabolism , Cell Wall/genetics , Cell Wall/metabolism , Female , Humans , Lactococcus lactis/chemistry , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Domains , Protein Structure, Secondary , Streptococcus agalactiae/genetics , Streptococcus agalactiae/metabolism , Structure-Activity Relationship
15.
J Appl Oral Sci ; 24(2): 126-35, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27119760

ABSTRACT

Objective Bacterial penetration of dentinal tubules via exposed dentine can lead to root caries and promote infections of the pulp and root canal system. The aim of this work was to develop a new experimental model for studying bacterial invasion of dentinal tubules within the human oral cavity. Material and Methods Sections of human root dentine were mounted into lower oral appliances that were worn by four human subjects for 15 d. Roots were then fixed, sectioned, stained and examined microscopically for evidence of bacterial invasion. Levels of invasion were expressed as Tubule Invasion Factor (TIF). DNA was extracted from root samples, subjected to polymerase chain reaction amplification of 16S rRNA genes, and invading bacteria were identified by comparison of sequences with GenBank database. Results All root dentine samples with patent tubules showed evidence of bacterial cell invasion (TIF value range from 5.7 to 9.0) to depths of 200 mm or more. A spectrum of Gram-positive and Gram-negative cell morphotypes were visualized, and molecular typing identified species of Granulicatella, Streptococcus, Klebsiella, Enterobacter, Acinetobacter, and Pseudomonas as dentinal tubule residents. Conclusion A novel in vivo model is described, which provides for human root dentine to be efficiently infected by oral microorganisms. A range of bacteria were able to initially invade dentinal tubules within exposed dentine. The model will be useful for testing the effectiveness of antiseptics, irrigants, and potential tubule occluding agents in preventing bacterial invasion of dentine.


Subject(s)
Bacteria/isolation & purification , Dental Pulp Cavity/microbiology , Dentin/microbiology , Tooth Root/microbiology , Biofilms , DNA, Bacterial , Dentin/ultrastructure , Humans , Polymerase Chain Reaction , RNA, Ribosomal, 16S , Reproducibility of Results , Surface Properties
16.
J. appl. oral sci ; 24(2): 126-135, Mar.-Apr. 2016. tab, graf
Article in English | LILACS | ID: lil-779909

ABSTRACT

ABSTRACT Objective Bacterial penetration of dentinal tubules via exposed dentine can lead to root caries and promote infections of the pulp and root canal system. The aim of this work was to develop a new experimental model for studying bacterial invasion of dentinal tubules within the human oral cavity. Material and Methods Sections of human root dentine were mounted into lower oral appliances that were worn by four human subjects for 15 d. Roots were then fixed, sectioned, stained and examined microscopically for evidence of bacterial invasion. Levels of invasion were expressed as Tubule Invasion Factor (TIF). DNA was extracted from root samples, subjected to polymerase chain reaction amplification of 16S rRNA genes, and invading bacteria were identified by comparison of sequences with GenBank database. Results All root dentine samples with patent tubules showed evidence of bacterial cell invasion (TIF value range from 5.7 to 9.0) to depths of 200 mm or more. A spectrum of Gram-positive and Gram-negative cell morphotypes were visualized, and molecular typing identified species of Granulicatella, Streptococcus, Klebsiella, Enterobacter, Acinetobacter, and Pseudomonas as dentinal tubule residents. Conclusion A novel in vivo model is described, which provides for human root dentine to be efficiently infected by oral microorganisms. A range of bacteria were able to initially invade dentinal tubules within exposed dentine. The model will be useful for testing the effectiveness of antiseptics, irrigants, and potential tubule occluding agents in preventing bacterial invasion of dentine.


Subject(s)
Humans , Bacteria/isolation & purification , Dental Pulp Cavity/microbiology , Dentin/microbiology , Tooth Root/microbiology , Biofilms , Dentin/ultrastructure , DNA, Bacterial , Polymerase Chain Reaction , Reproducibility of Results , RNA, Ribosomal, 16S , Surface Properties
17.
Pathog Dis ; 74(3)2016 Apr.
Article in English | MEDLINE | ID: mdl-26755532

ABSTRACT

Candida-associated stomatitis affects up to 60% of denture wearers, and Candida albicans remains the most commonly isolated fungal species. The oral bacteria Actinomyces oris and Streptococcus oralis are abundant in early dental plaque. The aims of this study were to determine the effects of S. oralis and A. oris on the development of C. albicans biofilms on denture material. Resin discs were coated with saliva and at early (1.5 h) or later (24 h) stages of biofilm development, cell numbers of each species were determined. Spatial distribution of microorganisms was visualized by confocal scanning laser microscopy of biofilms labelled by differential fluorescence or by fluorescence in situ hybridization. Interkingdom interactions underpinning biofilm development were also evaluated planktonically utilizing fluorescence microscopy. Synergistic interactions between all three species occurred within biofilms and planktonically. Bacterial cells coaggregated with each other and adhered singly or in coaggregates to C. albicans hyphal filaments. Streptococcus oralis appeared to enhance hyphal filament production and C. albicans biovolume was increased 2-fold. Concomitantly, cell numbers of S. oralis and A. oris were enhanced by C. albicans. Thus, cooperative physical and metabolic processes occurring between these three microbial species intensify pathogenic plaque communities on denture surfaces.


Subject(s)
Actinomyces/growth & development , Biofilms/growth & development , Candida albicans/growth & development , Dentures/microbiology , Streptococcus oralis/growth & development , Symbiosis/physiology , Adult , Humans , In Situ Hybridization, Fluorescence , Microbial Consortia/physiology , Microscopy, Confocal , Saliva , Stomatitis, Denture/microbiology
18.
Pathog Dis ; 74(3)2016 Apr.
Article in English | MEDLINE | ID: mdl-26772652

ABSTRACT

The fungus Candida albicans colonizes oral cavity surfaces and is carried by up to 60% of human populations. Biofilm development by C. albicans may be modulated by oral streptococci, such as Streptococcus gordonii, S. oralis or S. mutans, so as to augment pathogenicity. In this study we sought to determine if the cell wall-associated secreted aspartyl proteinase Sap9 was necessary for hyphal adhesin functions associated with biofilm community development. A sap9Δ mutant of C. albicans SC5314 formed biofilms that were flatter, and contained fewer blastospores and more hyphal filaments than the parent strain. This phenotypic difference was accentuated under flow (shear) conditions and in the presence of S. gordonii. Dual-species biofilms of C. albicans sap9Δ with S. oralis, S. sanguinis, S. parasanguinis, S. mutans and Enterococcus faecalis all contained more matted hyphae and more bacteria bound to substratum compared to C. albicans wild type. sap9Δ mutant hyphae showed significantly increased cell surface hydrophobicity, ∼25% increased levels of binding C. albicans cell wall protein Als3, and reduced interaction with Eap1, implicating Sap9 in fungal cell-cell recognition. These observations suggest that Sap9 is associated with protein-receptor interactions between fungal cells, and with interkingdom communication in the formation of polymicrobial biofilm communities.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Biofilms/growth & development , Candida albicans/metabolism , Enterococcus faecalis/metabolism , Fungal Proteins/metabolism , Streptococcus gordonii/metabolism , Streptococcus mutans/metabolism , Adult , Aspartic Acid Endopeptidases/genetics , Candida albicans/genetics , Candida albicans/pathogenicity , Fungal Proteins/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Hyphae/metabolism , Microbial Interactions/physiology , Microscopy, Confocal , Mouth/microbiology
19.
Cell Microbiol ; 18(6): 844-58, 2016 06.
Article in English | MEDLINE | ID: mdl-26639759

ABSTRACT

The oral anaerobe Porphyromonas gingivalis is associated with the development of cancers including oral squamous cell carcinoma (OSCC). Here, we show that infection of gingival epithelial cells with P. gingivalis induces expression and nuclear localization of the ZEB1 transcription factor, which controls epithelial-mesenchymal transition. P. gingivalis also caused an increase in ZEB1 expression as a dual species community with Fusobacterium nucleatum or Streptococcus gordonii. Increased ZEB1 expression was associated with elevated ZEB1 promoter activity and did not require suppression of the miR-200 family of microRNAs. P. gingivalis strains lacking the FimA fimbrial protein were attenuated in their ability to induce ZEB1 expression. ZEB1 levels correlated with an increase in expression of mesenchymal markers, including vimentin and MMP-9, and with enhanced migration of epithelial cells into matrigel. Knockdown of ZEB1 with siRNA prevented the P. gingivalis-induced increase in mesenchymal markers and epithelial cell migration. Oral infection of mice by P. gingivalis increased ZEB1 levels in gingival tissues, and intracellular P. gingivalis were detected by antibody staining in biopsy samples from OSCC. These findings indicate that FimA-driven ZEB1 expression could provide a mechanistic basis for a P. gingivalis contribution to OSCC.


Subject(s)
Gingiva/microbiology , Porphyromonas gingivalis/pathogenicity , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Cell Movement , Epithelial Cells/microbiology , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Fimbriae, Bacterial/metabolism , Gene Expression Regulation , Gingiva/cytology , Gingiva/metabolism , Host-Pathogen Interactions , Humans , Keratinocytes/microbiology , Keratinocytes/pathology , Mice, Inbred BALB C , MicroRNAs/genetics , Mouth Neoplasms/microbiology , Porphyromonas gingivalis/genetics , Promoter Regions, Genetic , Zinc Finger E-box-Binding Homeobox 1/genetics
20.
J Mater Sci Mater Med ; 26(6): 201, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26123234

ABSTRACT

Dental implants are an increasingly popular solution to missing teeth. Implants are prone to colonisation by pathogenic oral bacteria which can lead to inflammation, destruction of bone and ultimately implant failure. The aim of this study was to investigate the use of chlorhexidine (CHX) hexametaphosphate (HMP) nanoparticles (NPs) with a total CHX concentration equivalent to 5 mM as a coating for dental implants. The CHX HMP NPs had mean diameter 49 nm and composition was confirmed showing presence of both chlorine and phosphorus. The NPs formed micrometer-sized aggregated surface deposits on commercially pure grade II titanium substrates following immersion-coating for 30 s. When CHX HMP NP-coated titanium specimens were immersed in deionised water, sustained release of soluble CHX was observed, both in the absence and presence of a salivary pellicle, for the duration of the study (99 days) without reaching a plateau. Control specimens exposed to a solution of aqueous 25 µM CHX (equivalent to the residual aqueous CHX present with the NPs) did not exhibit CHX release. CHX HMP NP-coated surfaces exhibited antimicrobial efficacy against oral primary colonising bacterium Streptococcus gordonii within 8 h. The antimicrobial efficacy was greater in the presence of an acquired pellicle which is postulated to be due to retention of soluble CHX by the pellicle.


Subject(s)
Chlorhexidine/chemistry , Coated Materials, Biocompatible/chemistry , Dental Implants , Nanoparticles/chemistry , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/chemistry , Bacterial Adhesion/drug effects , Bacterial Load , Chlorhexidine/administration & dosage , Dental Implants/adverse effects , Dental Implants/microbiology , Humans , In Vitro Techniques , Materials Testing , Nanoparticles/administration & dosage , Phosphates/administration & dosage , Phosphates/chemistry , Streptococcus gordonii/drug effects , Streptococcus gordonii/physiology , Surface Properties , Titanium/adverse effects , Titanium/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...