Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Microbiol Spectr ; 7(4)2019 07.
Article in English | MEDLINE | ID: mdl-31267891

ABSTRACT

The human oral-nasal mucosa is the primary reservoir for Streptococcus pyogenes infections. Although the most common infection of consequence in temperate climates is pharyngitis, the past 25 years have witnessed a dramatic increase in invasive disease in many regions of the world. Historically, S. pyogenes has been associated with sepsis and fulminate systemic infections, but the mechanism by which these streptococci traverse mucosal or epidermal barriers is not understood. The discovery that S. pyogenes can be internalized by mammalian epithelial cells at high frequencies (1-3) and/or open tight junctions to pass between cells (4) provides potential explanations for changes in epidemiology and the ability of this species to breach such barriers. In this article, the invasins and pathways that S. pyogenes uses to reach the intracellular state are reviewed, and the relationship between intracellular invasion and human disease is discussed.


Subject(s)
Receptors, Cell Surface/metabolism , Streptococcal Infections/microbiology , Streptococcus pyogenes/physiology , Animals , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/microbiology , Humans , Pharyngitis/genetics , Pharyngitis/metabolism , Pharyngitis/microbiology , Receptors, Cell Surface/genetics , Streptococcal Infections/genetics , Streptococcal Infections/metabolism , Streptococcus pyogenes/genetics
2.
J Pediatric Infect Dis Soc ; 6(2): 187-196, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28204534

ABSTRACT

BACKGROUND.: Despite the significant burden of disease associated with infection by group A streptococcus (GAS), little is known about the human immune response to GAS antigens after natural infection. METHODS.: We evaluated 195 serum samples obtained prospectively over a consecutive 24-month period from 41 pediatric subjects who experienced a new pharyngeal GAS acquisition. An enzyme-linked immunoassay was used to determine the kinetics and antigen specificity of antibodies against 13 shared GAS antigens and 18 type-specific M peptides. The majority of the antigens tested are currently being considered as vaccine candidates. RESULTS.: Twelve M types of GAS were recovered from 41 subjects who experienced 51 new GAS acquisitions that elicited antibody responses against at least 1 of the 31 antigens tested (immunologically significant new GAS acquisitions). The immune responses to the 13 shared antigens were highly variable. Increases in antibody levels were detected against a mean of 3.5 shared antigens (range, 1-8). Antibody responses to the homologous M peptide were observed in 32 (63%) of the 51 episodes. Seven subjects acquired more than 1 M type of GAS. There were no new immunologically significant acquisitions of an M type against which the subject had preexisting antibodies to the homologous M peptide. Of the subjects with new GAS acquisition, 65% were asymptomatic, yet immune responses were detected against 1 or more GAS antigens. Immune responses to streptolysin O and/or deoxyribonuclease B were observed after 67% of the new GAS acquisitions. Persistently positive (>12 weeks) throat culture results were returned for 20% of the 41 subjects despite immune responses to homologous M peptides and/or shared antigens. CONCLUSIONS.: The availability of throat culture results, GAS isolates, and serial serum samples collected prospectively over a 2-year period of observation provided a unique opportunity for us to assess the serologic status of pediatric subjects before and after new pharyngeal acquisitions of GAS. With the exception of antibody responses to the homologous M peptides, no clear pattern of immune responses against the remaining GAS antigens was seen. There were no new immunologically significant acquisitions of emm types of GAS against which the subjects had preexisting elevated levels of antibodies against the homologous M peptide. The observation that 65% of new GAS acquisitions caused no symptoms yet were immunologically significant suggests that the majority of infections are not detected, which would result in missed opportunities for primary prevention of rheumatic fever and rheumatic heart disease with appropriate antimicrobial therapy.


Subject(s)
Pharyngeal Diseases/microbiology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Adolescent , Antibodies, Bacterial/immunology , Antibody Formation/immunology , Antigens, Bacterial/immunology , Child , Enzyme-Linked Immunosorbent Assay , Humans , Longitudinal Studies , Pharyngeal Diseases/immunology , Prospective Studies , Streptococcal Infections/microbiology
3.
J Clin Invest ; 126(1): 303-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26657857

ABSTRACT

Group A streptococcal (GAS) infection induces the production of Abs that cross-react with host neuronal proteins, and these anti-GAS mimetic Abs are associated with autoimmune diseases of the CNS. However, the mechanisms that allow these Abs to cross the blood-brain barrier (BBB) and induce neuropathology remain unresolved. We have previously shown that GAS infection in mouse models induces a robust Th17 response in nasal-associated lymphoid tissue (NALT). Here, we identified GAS-specific Th17 cells in tonsils of humans naturally exposed to GAS, prompting us to explore whether GAS-specific CD4+ T cells home to mouse brains following i.n. infection. Intranasal challenge of repeatedly GAS-inoculated mice promoted migration of GAS-specific Th17 cells from NALT into the brain, BBB breakdown, serum IgG deposition, microglial activation, and loss of excitatory synaptic proteins under conditions in which no viable bacteria were detected in CNS tissue. CD4+ T cells were predominantly located in the olfactory bulb (OB) and in other brain regions that receive direct input from the OB. Together, these findings provide insight into the immunopathology of neuropsychiatric complications that are associated with GAS infections and suggest that crosstalk between the CNS and cellular immunity may be a general mechanism by which infectious agents exacerbate symptoms associated with other CNS autoimmune disorders.


Subject(s)
Brain/pathology , Palatine Tonsil/microbiology , Streptococcus pyogenes/immunology , Th17 Cells/physiology , Animals , Blood-Brain Barrier , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Female , Immunoglobulin G/blood , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Mice , Mice, Inbred C57BL , Tight Junctions/physiology
4.
PLoS One ; 10(6): e0128862, 2015.
Article in English | MEDLINE | ID: mdl-26067103

ABSTRACT

The invention of peptide-MHC-tetramer technology to label antigen-specific T cells has led to an enhanced understanding of T lymphocyte biology. Here we describe the development of an in situ pMHC-II tetramer staining method to visualize antigen-specific CD4+ T cells in tissues. This method complements other methods developed that similarly use MHC class II reagents to stain antigen-specific CD4+ T cells in situ. In this study, we used group A streptococcus (GAS) expressing a surrogate peptide (2W) to inoculate C57BL/6 mice, and used fresh nasal-associated lymphoid tissues (NALT) in optimizing the in situ staining of 2W:I-Ab specific CD4+ T cells. The results showed 2W:I-Ab tetramer-binding CD4+ T cells in GAS-2W but not GAS infected mice. This method holds promise to be broadly applicable to study the localization, abundance, and phenotype of antigen-specific CD4+ T cells in undisrupted tissues.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Peptides/immunology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/metabolism , Female , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Peptides/chemistry , Peptides/metabolism , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcal Infections/veterinary , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/physiology
5.
PLoS Pathog ; 10(5): e1004155, 2014 May.
Article in English | MEDLINE | ID: mdl-24875883

ABSTRACT

Establishing the genetic determinants of niche adaptation by microbial pathogens to specific hosts is important for the management and control of infectious disease. Streptococcus pyogenes is a globally prominent human-specific bacterial pathogen that secretes superantigens (SAgs) as 'trademark' virulence factors. SAgs function to force the activation of T lymphocytes through direct binding to lateral surfaces of T cell receptors and class II major histocompatibility complex (MHC-II) molecules. S. pyogenes invariably encodes multiple SAgs, often within putative mobile genetic elements, and although SAgs are documented virulence factors for diseases such as scarlet fever and the streptococcal toxic shock syndrome (STSS), how these exotoxins contribute to the fitness and evolution of S. pyogenes is unknown. Here we show that acute infection in the nasopharynx is dependent upon both bacterial SAgs and host MHC-II molecules. S. pyogenes was rapidly cleared from the nasal cavity of wild-type C57BL/6 (B6) mice, whereas infection was enhanced up to ∼10,000-fold in B6 mice that express human MHC-II. This phenotype required the SpeA superantigen, and vaccination with an MHC -II binding mutant toxoid of SpeA dramatically inhibited infection. Our findings indicate that streptococcal SAgs are critical for the establishment of nasopharyngeal infection, thus providing an explanation as to why S. pyogenes produces these potent toxins. This work also highlights that SAg redundancy exists to avoid host anti-SAg humoral immune responses and to potentially overcome host MHC-II polymorphisms.


Subject(s)
Bacterial Proteins/metabolism , Exotoxins/metabolism , Histocompatibility Antigens Class II/immunology , Membrane Proteins/metabolism , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Superantigens/immunology , Acute Disease , Animals , Bacterial Proteins/immunology , Exotoxins/immunology , Humans , Membrane Proteins/immunology , Mice, Inbred C57BL , Nasopharynx/immunology , Receptors, Antigen, T-Cell/immunology , Streptococcal Infections/prevention & control , Streptococcus pyogenes/genetics , Superantigens/genetics , T-Lymphocytes/immunology
6.
Proc Natl Acad Sci U S A ; 107(13): 5937-42, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20231435

ABSTRACT

Recurrent group A Streptococcus (GAS) tonsillitis and associated autoimmune diseases indicate that the immune response to this organism can be ineffective and pathological. TGF-beta1 is recognized as an essential signal for generation of regulatory T cells (Tregs) and T helper (Th) 17 cells. Here, the impact of TGF-beta1 induction on the T-cell response in mouse nasal-associated lymphoid tissue (NALT) following intranasal (i.n.) infections is investigated. ELISA and TGF-beta1-luciferase reporter assays indicated that persistent infection of mouse NALT with GAS sets the stage for TGF-beta1 and IL-6 production, signals required for promotion of a Th17 immune response. As predicted, IL-17, the Th17 signature cytokine, was induced in a TGF-beta1 signaling-dependent manner in single-cell suspensions of both human tonsils and NALT. Intracellular cytokine staining and flow cytometry demonstrated that CD4(+) IL-17(+) T cells are the dominant T cells induced in NALT by i.n. infections. Moreover, naive mice acquired the potential to clear GAS by adoptive transfer of CD4(+) T cells from immunized IL-17A(+)/(+) mice but not cells from IL-17A(-)/(-) mice. These experiments link specific induction of TGF-beta1 by a bacterial infection to an in vivo Th17 immune response and show that this cellular response is sufficient for protection against GAS. The association of a Th17 response with GAS infection reveals a potential mechanism for destructive autoimmune responses in humans.


Subject(s)
Interleukin-17/metabolism , Streptococcal Infections/immunology , Streptococcal Infections/pathology , Streptococcus pyogenes , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Transforming Growth Factor beta1/biosynthesis , Animals , Cell Differentiation/immunology , Cytokines/biosynthesis , Female , Humans , Immunity, Cellular , Interleukin-17/deficiency , Interleukin-17/genetics , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Listeriosis/pathology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nasal Cavity/immunology , Nasal Cavity/pathology , Signal Transduction/immunology , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Th1 Cells/immunology , Th1 Cells/pathology , Tonsillitis/immunology , Tonsillitis/pathology
7.
Nat Immunol ; 11(1): 83-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19935657

ABSTRACT

We used a sensitive method based on tetramers of peptide and major histocompatibility complex II (pMHCII) to determine whether CD4(+) memory T cells resemble the T helper type 1 (T(H)1) and interleukin 17 (IL-17)-producing T helper (T(H)17) subsets described in vitro. Intravenous or intranasal infection with Listeria monocytogenes induced pMHCII-specific CD4(+) naive T cells to proliferate and produce effector cells, about 10% of which resembled T(H)1 or T(H)17 cells, respectively. T(H)1 cells were also present among the memory cells that survived 3 months after infection, whereas T(H)17 cells disappeared. The short lifespan of T(H)17 cells was associated with small amounts of the antiapoptotic protein Bcl-2, the IL-15 receptor and the receptor CD27, and little homeostatic proliferation. These results suggest that T(H)1 cells induced by intravenous infection are more efficient at entering the memory pool than are T(H)17 cells induced by intranasal infection.


Subject(s)
Interleukin-17/immunology , Listeriosis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Cell Line , Cell Proliferation , Cell Survival/immunology , Clone Cells/immunology , Clone Cells/pathology , Flow Cytometry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Host-Pathogen Interactions/immunology , Immunologic Memory/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/physiology , Listeriosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Interleukin-15/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , Th1 Cells/metabolism , Th1 Cells/pathology , Time Factors , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
8.
FEMS Immunol Med Microbiol ; 55(3): 422-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19243434

ABSTRACT

Streptococcus pyogenes is a major causative agent of tonsillitis or pharyngitis in children. Streptococcus pyogenes can persist in tonsils, and one-third of children treated with antibiotics continue to shed streptococci and have recurrent infections. Mouse nasal-associated lymphoid tissue (NALT) is functionally analogous to human oropharyngeal lymphoid tissues, and serves as a model for characterization of the mucosal innate immune response to S. pyogenes. Wild-type S. pyogenes induces transcription of both type I and interferon-gamma (IFN-gamma)-responsive genes, proinflammatory genes and acute-phase response proteins 24 h after intranasal infection. Invasion of NALT and the induction of the interferon response were not dependent on expression of antiphagocytic M protein. Intranasal infection induces a substantial influx of neutrophils into NALT at 24 h, which declines by 48 h after infection. Infection of IFN-gamma(-/-) [IFN-gamma knock-out mouse (GKO)] C57BL/6 mice with wild-type S. pyogenes resulted in local dissemination of bacteria to draining lymph nodes (LN), but did not lead to systemic infection by 48 h after infection. Infected GKO mice had an increased influx of neutrophils into NALT compared with immunocompetent mice. Thus, IFN-gamma-induced responses are required to prevent local dissemination of streptococci to the draining LN.


Subject(s)
Interferons/biosynthesis , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Streptococcus pyogenes/immunology , Animals , Cell Survival , Colony Count, Microbial , Female , Gene Expression Profiling , Interferons/deficiency , Lymph Nodes/microbiology , Lymphoid Tissue/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/microbiology , Neutrophils/immunology
9.
Infect Immun ; 75(6): 3188-91, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17371855

ABSTRACT

Streptococcus pyogenes is a major cause of pharyngitis in humans and encodes several fibronectin-binding proteins. M protein and protein F1 (PrtF1/SfbI) are differentially regulated by CO(2) and O(2), respectively, and both mediate the invasion of epithelial cells. This study examined whether PrtF1/SfbI shares other properties with M protein. Expression of the PrtF1/SfbI protein by an M-negative mutant conferred resistance to phagocytosis and partial inhibition of C3 deposition on the S. pyogenes surface.


Subject(s)
Adhesins, Bacterial/metabolism , Epithelial Cells/microbiology , Phagocytosis/physiology , Streptococcus pyogenes/metabolism , Adhesins, Bacterial/genetics , Cell Line , Humans
10.
Cell Microbiol ; 9(6): 1519-28, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17298394

ABSTRACT

Efficient group A streptococcus (GAS) invasion of mammalian cells requires fibronectin (Fn) binding proteins, such as M1 and PrtF1/SfbI, that bridge bacteria to integrins and activate cellular signalling for ingestion. Previous studies of GAS invasion, mediated by both proteins, suggest a common signalling pathway. However, distinct cellular morphological changes at the port of bacterial entry suggest that different signals are also induced. Here we report that paxillin is phosphorylated in response to Fn-bound GAS that express either M1 or PrtF1/SfbI protein, but is not phosphorylated in response to a mutant deficient in both proteins. Inhibition of paxillin phosphorylation by a tyrosine kinase inhibitor, PP2, or by expression of a dominant negative form of paxillin significantly reduced invasion by M1+ but did not affect ingestion of PrtF1/SfbI+ strains. In contrast, another tyrosine inhibitor, genistein, did not significantly prevent paxillin phosphorylation and had no effect on ingestion of the M1+ strain, but reduced PrtF1/SfbI-mediated entry. This suggests that paxillin phosphorylation is induced by both proteins but only required for M1-mediated invasion. A bifurcation point, downstream of integrin-linked kinase (ILK) and phosphoinositide 3-kinase, likely accounts for the distinct morphological changes. Furthermore, ILK activity is indispensable for M1-induced paxillin recruitment and phosphorylation.


Subject(s)
Paxillin/metabolism , Protein Serine-Threonine Kinases/metabolism , Streptococcus pyogenes/pathogenicity , Adhesins, Bacterial/metabolism , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Focal Adhesions/metabolism , HeLa Cells , Humans , Lactococcus/metabolism , Phosphorylation , Pyrimidines/pharmacology , Signal Transduction , Streptococcus pyogenes/metabolism
12.
Cell Microbiol ; 8(2): 257-66, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16441436

ABSTRACT

Entry of Streptococcus pyogenes or group A streptococcus (GAS) into host cells is mediated by fibronectin bound to surface proteins, M1 or PrtF1, forming a bridge to alpha5beta1 integrins. This interaction leads to cytoskeletal rearrangement and uptake of streptococci. We postulated that integrin-linked kinase (ILK), which directly associates with integrins, is the universal link between integrins and several bacterial pathogens. We showed that inhibition of ILK expression by siRNA silencing, or ILK kinase activity by chemical inhibitors or expression of a dominant negative form of ILK reduced M1-mediated invasion of epithelial cells up to 80%. To evaluate the ILK requirement for PrtF1-mediated GAS invasion, a M1-PrtF1+ recombinant strain within the M1 background was constructed. Inhibition of ILK kinase activity also significantly reduced invasion of epithelial cells by this recombinant and wild-type strain JRS4 that expresses PrtF1. In addition, impaired ILK kinase activity results in significant reduction of integrin-dependent invasion mediated by invasins of two other important pathogens, Staphylococcus aureus and Yersinia spp. This study suggests that bacterial pathogens evolved different molecules and strategies to exploit the host integrin signalling pathway for their survival.


Subject(s)
Epithelial Cells/microbiology , Integrins/metabolism , Protein Serine-Threonine Kinases/metabolism , Streptococcus pyogenes/physiology , Yersinia/physiology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Streptococcus pyogenes/genetics
13.
Proc Natl Acad Sci U S A ; 102(51): 18391-6, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16344483

ABSTRACT

The structure of a cell surface enzyme from a gram-positive pathogen has been determined to 2-A resolution. Gram-positive pathogens have a thick cell wall to which proteins and carbohydrate are covalently attached. Streptococcal C5a peptidase (SCP), is a highly specific protease and adhesin/invasin. Structural analysis of a 949-residue fragment of the [D130A,S512A] mutant of SCP from group B Streptococcus (S. agalactiae, SCPB) revealed SCPB is composed of five distinct domains. The N-terminal subtilisin-like protease domain has a 134-residue protease-associated domain inserted into a loop between two beta-strands. This domain also contains one of two Arg-Gly-Asp (RGD) sequences found in SCPB. At the C terminus are three fibronectin type III (Fn) domains. The second RGD sequence is located between Fn1 and Fn2. Our analysis suggests that SCP binding to integrins by the RGD motifs may stabilize conformational changes required for substrate binding.


Subject(s)
Adhesins, Bacterial/chemistry , Cell Wall/enzymology , Endopeptidases/chemistry , Streptococcus agalactiae/enzymology , Adhesins, Bacterial/metabolism , Amino Acid Motifs , Amino Acid Sequence , Crystallography, X-Ray , Endopeptidases/metabolism , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary
14.
Infect Immun ; 73(12): 7878-86, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16299278

ABSTRACT

C5a peptidase, also called SCPA (surface-bound C5a peptidase), is a surface-bound protein on group A streptococci (GAS), etiologic agents for a variety of human diseases including pharyngitis, impetigo, toxic shock, and necrotizing fasciitis, as well as the postinfection sequelae rheumatic fever and rheumatic heart disease. This protein is highly conserved among different serotypes and is also expressed in human isolates of group B, C, and G streptococci. Human tonsils are the primary reservoirs for GAS, maintaining endemic disease across the globe. We recently reported that GAS preferentially target nasal mucosa-associated lymphoid tissue (NALT) in mice, a tissue functionally analogous to human tonsils. Experiments using a C5a peptidase loss-of-function mutant and an intranasal infection model showed that this protease is required for efficient colonization of NALT. An effective vaccine should prevent infection of this secondary lymphoid tissue; therefore, the potential of anti-SCPA antibodies to protect against streptococcal infection of NALT was investigated. Experiments showed that GAS colonization of NALT was significantly reduced following intranasal immunization of mice with recombinant SCPA protein administered alone or with cholera toxin, whereas a high degree of GAS colonization of NALT was observed in control mice immunized with phosphate-buffered saline only. Moreover, administration of anti-SCPA serum by the intranasal route protected mice against streptococcal infection. These results suggest that intranasal immunization with SCPA would prevent colonization and infection of human tonsils, thereby eliminating potential reservoirs that maintain endemic disease.


Subject(s)
Adhesins, Bacterial/immunology , Endopeptidases/immunology , Immunization, Passive , Nasal Mucosa/microbiology , Streptococcal Infections/prevention & control , Streptococcal Vaccines , Streptococcus pyogenes/immunology , Adhesins, Bacterial/administration & dosage , Adhesins, Bacterial/genetics , Administration, Intranasal , Animals , Cyclohexanecarboxylic Acids/blood , Endopeptidases/administration & dosage , Endopeptidases/genetics , Female , Humans , Immune Sera/administration & dosage , Immune Sera/immunology , Immunoglobulin A/blood , Lymphoid Tissue/microbiology , Mice , Mice, Inbred BALB C , Palatine Tonsil/microbiology , Rabbits , Streptococcal Vaccines/administration & dosage
15.
Clin Infect Dis ; 41(8): 1150-6, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16163634

ABSTRACT

Infections due to group A streptococci (GAS) represent a public health problem of major proportions in both developing and developed countries. Currently available methods of prevention are either inadequate or ineffective, as attested to by the morbidity and mortality associated with this ubiquitous pathogen worldwide. Advances in molecular biology have shed new light on the pathogenesis of GAS infections and have identified a number of virulence factors as potential vaccine targets. Therefore, the National Institute of Allergy and Infectious Diseases convened an expert workshop in March 2004 to review the available data and to explore the microbiologic, immunologic, epidemiologic, and economic issues involved in development and implementation of a safe and effective GAS vaccine. Participants included scientists and clinicians involved in GAS research, as well as representatives of United States federal agencies (Centers for Disease Control and Prevention, Food and Drug Administration, Department of Defense, and National Institute of Allergy and Infectious Diseases), the World Health Organization, and the pharmaceutical industry. This report summarizes the deliberations of the workshop.


Subject(s)
Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Developed Countries , Developing Countries , National Institutes of Health (U.S.) , Streptococcal Infections/epidemiology , Streptococcus pyogenes , United States
16.
Cell Microbiol ; 7(5): 645-53, 2005 May.
Article in English | MEDLINE | ID: mdl-15839894

ABSTRACT

Membrane cofactor protein (MCP or CD46), a widely distributed complement regulatory human protein, is a cell surface receptor for many pathogens including group A streptococci (GAS). The surface M protein of GAS binds CD46 and mediates GAS adherence to keratinocytes. In the present study, we studied the role of CD46 in GAS invasion of human lung epithelial cells, A549. Anti-CD46 antibody which specifically blocks the domain to which M protein binds inhibited adherence to and invasion of A549 cells by GAS. Moreover, downregulation of CD46 expression on A549 by RNA interference resulted in reduced invasion of these cells by GAS. A mutant form of CD46 with a deletion in the cytoplasmic domain was overexpressed in A549 cells, which resulted in partial inhibition of invasion. This indicates that the cytoplasmic tail is required for CD46 to promote invasion by GAS. Invasion assays with Lactococcus lactis that express M protein demonstrated the dependence of CD46-promoted invasion on interaction with M protein. In addition, CD46-mediated invasion was also found to be dependent on the extracellular matrix protein fibronectin.


Subject(s)
Epithelial Cells/microbiology , Integrin alpha5beta1/biosynthesis , Membrane Cofactor Protein/biosynthesis , Streptococcus pyogenes/physiology , Antigens, Bacterial/metabolism , Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Cell Line , Down-Regulation , Epithelial Cells/metabolism , Fibronectins/metabolism , Flow Cytometry , Humans , Lactococcus lactis/metabolism , RNA Interference , Streptococcus pyogenes/pathogenicity
17.
Infect Immun ; 73(2): 859-64, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15664926

ABSTRACT

The streptococcal pyrogenic exotoxin B (SpeB) is an important virulence factor of group A streptococci (GAS) with cysteine protease activity. Maturation of SpeB to a proteolytically active form was suggested to be dependent on cell-wall-anchored M1 protein, the major surface protein of GAS (M. Collin and A. Olsen, Mol. Microbiol. 36:1306-1318, 2000). Collin and Olsen showed that mutant GAS strains expressing truncated M protein secrete a conformationally different form of unprocessed SpeB with no proteolytic activity. Alternatively, we hypothesized that a truncated M protein may interfere with processing of this secreted protease, and therefore we tested cysteine protease activity in genetically defined mutant strains that express either no M protein or membrane-anchored M protein with an in-frame deletion of the AB repeat region. Measurements of SpeB activity by cleavage of a substrate n-benzoyl-Pro-Phe-Arg-p-nitroanilide hydrochloride showed that the proteolytic activities in culture supernatants of both mutants were similar to those from the wild-type strain. In addition, Western blot analysis of culture supernatants showed that SpeB expression and processing to a mature form was unaffected by either deletion mutation. Therefore, we conclude that M protein is not required for maturation of the streptococcal cysteine protease SpeB.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Cysteine Endopeptidases/metabolism , Exotoxins/metabolism , Streptococcus pyogenes/metabolism , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/genetics , Cell Culture Techniques , Mutation , Streptococcus pyogenes/genetics
18.
Vaccine ; 22(31-32): 4332-41, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15474726

ABSTRACT

Group A streptococci (S. pyogenes) are responsible for pharyngitis, impetigo and several more serious diseases. Emergence of toxic shock, and necrotizing fasciitis, associated with this pathogen over the past 10 years, has generated interest in development of a vaccine, which would prevent infections and potential serious complications. The highly conserved C5a peptidase that is expressed on the surface of group A streptococcus and other streptococcal species, associated with human infections, is a prime vaccine candidate. Here, we report construction of an inactive form of the peptidase and test its potential to induce protection in mice from intranasal challenge with either serotype M1 and M49 strains of streptococci. Mice were immunized by subcutaneous administration of recombinant proteins, mixed with Alum and monophosphoryl lipid A (MPL) adjuvants. Control mice were vaccinated with tetanus toxoid in the same adjuvants. Preparations of SCPA protein were highly immunogenic in mice. Antibody directed against protein from either group A (SCPAw) or group B (SCPBw) streptococci neutralized activity associated with both enzymes. Streptococci were cleared from the oral-nasal mucosa of mice immunized with vaccine protein more rapidly than those immunized with tetanus toxoid. Moreover, immunization with either protein enhanced clearance of group A streptococci from the lung. These results suggest that parenteral vaccination with SCPBw protein will provide protection against infection by either group A or B streptococci.


Subject(s)
Adhesins, Bacterial/immunology , Endopeptidases/immunology , Nasal Cavity/microbiology , Streptococcus agalactiae/immunology , Streptococcus pyogenes/immunology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Adjuvants, Immunologic , Amino Acid Substitution , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/genetics , Cell Line , Endopeptidases/genetics , Endopeptidases/metabolism , Enzyme-Linked Immunosorbent Assay , Fibronectins/metabolism , Immunization , Injections, Subcutaneous , Lung/immunology , Lung/microbiology , Mice , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Neutralization Tests , Oropharynx/microbiology , Phagocytosis/immunology , Protein Binding , Vaccines, Synthetic/immunology
19.
Eur J Immunol ; 34(10): 2843-53, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15368301

ABSTRACT

CD4 T cells are important for development of long-term immunity to bacterial infections. Here we describe construction of a group A streptococcus (GAS) strain that expresses the model ovalbumin epitope (OVA) on its surface, and the use of this strain in adoptive transfer experiments to study CD4 T cell response to bacterial infection in nasal-associated lymphoid tissue (NALT), which was previously shown to be a specific target for GAS colonization. The OVA(+) GAS, but not the wild-type strain was shown to activate CD4 T cells in an antigen-specific manner both in vitro and in vivo. After intranasal infection of mice with this strain, OVA-specific CD4 T cells were first activated in NALT, which is functionally equivalent to human tonsils, rather than in the cervical lymph nodes. During localized infection, OVA(+) GAS induced rapid and prolonged activation of CD4 T cells at higher magnitudes in the NALT than in draining lymph nodes and spleen, where CD4 T cells underwent little or no activation. In contrast, systemic infection induced significantly higher activation of CD4 T cells in both lymph nodes and spleens, compared to when the infection was localized in NALT. Further investigation of cellular immune responses in NALT during GAS infection using adoptive T cell transfer, combined with the model antigen on the pathogen may ultimately shed light on mechanisms for failure of children to develop protective immune responses following streptococcal tonsillitis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Lymphoid Tissue/immunology , Nose/immunology , Streptococcal Infections/immunology , Adoptive Transfer , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/immunology , Carrier Proteins/metabolism , Disease Models, Animal , Flow Cytometry , Mice , Ovalbumin/immunology , Ovalbumin/metabolism , Polymerase Chain Reaction , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Streptococcus pyogenes/immunology
20.
Indian J Med Res ; 119 Suppl: 44-7, 2004 May.
Article in English | MEDLINE | ID: mdl-15232161

ABSTRACT

BACKGROUND & OBJECTIVES: Group A Streptococcus, causative agent of several clinical manifestations codes for multiple protein invasins which help the bacterium to enter non-phagocytic cells. C5a peptidase (SCPA) is a surface protein conserved among different serotypes of M1 strain. The present study was taken up to study SCPA promoted fibronectin independent entry of GAS into epithelial cells. METHODS: An isogenic 90226 emm1deltaAB (M1(-)) mutant was constructed with thermosensitive pGhost vector. This isogenic M1(-) mutant expressed SCPA on the surface as determined by Western blotting and immunofluorescence. RESULTS: On preincubation with anti-SCPA serum, the isogenic M1(-) strain exhibited 54 per cent decreased invasion as compared to the bacteria incubated with control serum. Also, purified recombinant SCPA proteins blocked internalization of M1(-) streptococci into HEp-2 cells. The M1(-) strain invaded at the same efficiency in the presence or absence of fibronectin. INTERPRETATION & CONCLUSION: These results suggested that SCPA acted as a potential invasin of group A streptococcus and promoted invasion independent of fibronectin.


Subject(s)
Adhesins, Bacterial/metabolism , Endopeptidases/metabolism , Epithelial Cells/microbiology , Fibronectins/physiology , Streptococcus pyogenes/physiology , Blotting, Western , Cell Line , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...