Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 11(3): e0411222, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37036369

ABSTRACT

The classical Bordetella species infect the respiratory tract of mammals. While B. bronchiseptica causes rather chronic respiratory infections in a variety of mammals, the human-adapted species B. pertussis and B. parapertussisHU cause an acute respiratory disease known as whooping cough or pertussis. The virulence factors include a type III secretion system (T3SS) that translocates effectors BteA and BopN into host cells. However, the regulatory mechanisms underlying the secretion and translocation activity of T3SS in bordetellae are largely unknown. We have solved the crystal structure of BopN of B. pertussis and show that it is similar to the structures of gatekeepers that control access to the T3SS channel from the bacterial cytoplasm. We further found that BopN accumulates at the cell periphery at physiological concentrations of calcium ions (2 mM) that inhibit the secretion of BteA and BopN. Deletion of the bopN gene in B. bronchiseptica increased secretion of the BteA effector into calcium-rich medium but had no effect on secretion of the T3SS translocon components BopD and BopB. Moreover, the ΔbopN mutant secreted approximately 10-fold higher amounts of BteA into the medium of infected cells than the wild-type bacteria, but it translocated lower amounts of BteA into the host cell cytoplasm. These data demonstrate that BopN is a Bordetella T3SS gatekeeper required for regulated and targeted translocation of the BteA effector through the T3SS injectisome into host cells. IMPORTANCE The T3SS is utilized by many Gram-negative bacteria to deliver effector proteins from bacterial cytosol directly into infected host cell cytoplasm in a regulated and targeted manner. Pathogenic bordetellae use the T3SS to inject the BteA and BopN proteins into infected cells and upregulate the production of the anti-inflammatory cytokine interleukin-10 (IL-10) to evade host immunity. Previous studies proposed that BopN acted as an effector in host cells. In this study, we report that BopN is a T3SS gatekeeper that regulates the secretion and translocation activity of Bordetella T3SS.


Subject(s)
Type III Secretion Systems , Whooping Cough , Animals , Humans , Type III Secretion Systems/metabolism , Calcium , Bordetella pertussis/metabolism , Virulence Factors/metabolism , Bacterial Proteins/metabolism , Mammals
2.
J Biol Chem ; 296: 100607, 2021.
Article in English | MEDLINE | ID: mdl-33789161

ABSTRACT

The respiratory pathogens Bordetella pertussis and Bordetella bronchiseptica employ a type III secretion system (T3SS) to inject a 69-kDa BteA effector protein into host cells. This effector is known to contain two functional domains, including an N-terminal lipid raft targeting (LRT) domain and a cytotoxic C-terminal domain that induces nonapoptotic and caspase-1-independent host cell death. However, the exact molecular mechanisms underlying the interaction of BteA with plasma membrane (PM) as well as its cytotoxic activity in the course of Bordetella infections remain poorly understood. Using a protein-lipid overlay assay and surface plasmon resonance, we show here that the recombinant LRT domain binds negatively charged membrane phospholipids. Specifically, we determined that the dissociation constants of the LRT domain-binding liposomes containing phosphatidylinositol 4,5-bisphosphate, phosphatidic acid, and phosphatidylserine were ∼450 nM, ∼490 nM, and ∼1.2 µM, respectively. Both phosphatidylserine and phosphatidylinositol 4,5-bisphosphate were required to target the LRT domain and/or full-length BteA to the PM of yeast cells. The membrane association further involved electrostatic and hydrophobic interactions of LRT and depended on a leucine residue in the L1 loop between the first two helices of the four-helix bundle. Importantly, charge-reversal substitutions within the L1 region disrupted PM localization of the BteA effector without hampering its cytotoxic activity during B. bronchiseptica infection of HeLa cells. The LRT-mediated targeting of BteA to the cytosolic leaflet of the PM of host cells is, therefore, dispensable for effector cytotoxicity.


Subject(s)
Bacterial Proteins/metabolism , Bordetella bronchiseptica/metabolism , Cell Membrane/metabolism , Lipid Bilayers/metabolism , Membrane Microdomains/metabolism , Phospholipids/metabolism , Bacterial Proteins/genetics , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/growth & development , HeLa Cells , Humans , Protein Binding , Protein Domains
3.
Article in English | MEDLINE | ID: mdl-33014891

ABSTRACT

Pertussis, also known as whooping cough, is a resurging acute respiratory disease of humans primarily caused by the Gram-negative coccobacilli Bordetella pertussis, and less commonly by the human-adapted lineage of B. parapertussisHU. The ovine-adapted lineage of B. parapertussisOV infects only sheep, while B. bronchiseptica causes chronic and often asymptomatic respiratory infections in a broad range of mammals but rarely in humans. A largely overlapping set of virulence factors inflicts the pathogenicity of these bordetellae. Their genomes also harbor a pathogenicity island, named bsc locus, that encodes components of the type III secretion injectosome, and adjacent btr locus with the type III regulatory proteins. The Bsc injectosome of bordetellae translocates the cytotoxic BteA effector protein, also referred to as BopC, into the cells of the mammalian hosts. While the role of type III secretion activity in the persistent colonization of the lower respiratory tract by B. bronchiseptica is well recognized, the functionality of the type III secretion injectosome in B. pertussis was overlooked for many years due to the adaptation of laboratory-passaged B. pertussis strains. This review highlights the current knowledge of the type III secretion system in the so-called classical Bordetella species, comprising B. pertussis, B. parapertussis, and B. bronchiseptica, and discusses its functional divergence. Comparison with other well-studied bacterial injectosomes, regulation of the type III secretion on the transcriptional and post-transcriptional level, and activities of BteA effector protein and BopN protein, homologous to the type III secretion gatekeepers, are addressed.


Subject(s)
Bordetella Infections , Bordetella bronchiseptica , Animals , Bacterial Proteins/genetics , Bordetella pertussis/genetics , Sheep , Type III Secretion Systems/genetics , Virulence Factors/genetics
4.
PLoS Pathog ; 16(8): e1008512, 2020 08.
Article in English | MEDLINE | ID: mdl-32776984

ABSTRACT

Bordetella bronchiseptica and Bordetella pertussis are closely related respiratory pathogens that evolved from a common bacterial ancestor. While B. bronchiseptica has an environmental reservoir and mostly establishes chronic infections in a broad range of mammals, B. pertussis is a human-specific pathogen causing acute pulmonary pertussis in infants and whooping cough illness in older humans. Both species employ a type III secretion system (T3SS) to inject a cytotoxic BteA effector protein into host cells. However, compared to the high BteA-mediated cytotoxicity of B. bronchiseptica, the cytotoxicity induced by B. pertussis BteA (Bp BteA) appears to be quite low and this has been attributed to the reduced T3SS gene expression in B. pertussis. We show that the presence of an alanine residue inserted at position 503 (A503) of Bp BteA accounts for its strongly attenuated cytotoxic potency. The deletion of A503 from Bp BteA greatly enhanced the cytotoxic activity of B. pertussis B1917 on mammalian HeLa cells and expression of Bp BteAΔA503 was highly toxic to Saccharomyces cerevisiae cells. Vice versa, insertion of A503 into B. bronchiseptica BteA (Bb BteA) strongly decreased its cytotoxicity to yeast and HeLa cells. Moreover, the production of Bp BteAΔA503 increased virulence of B. pertussis B1917 in the mouse model of intranasal infection (reduced LD50) but yielded less inflammatory pathology in infected mouse lungs at sublethal infectious doses. This suggests that A503 insertion in the T3SS effector Bp BteA may represent an evolutionary adaptation that fine-tunes B. pertussis virulence and host immune response.


Subject(s)
Alanine/metabolism , Bacterial Proteins/metabolism , Bordetella pertussis/physiology , Gene Expression Regulation, Bacterial , Whooping Cough/pathology , Alanine/genetics , Animals , Bacterial Proteins/genetics , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mutation , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Virulence , Whooping Cough/genetics , Whooping Cough/microbiology
5.
Nat Microbiol ; 3(10): 1122-1130, 2018 10.
Article in English | MEDLINE | ID: mdl-30224799

ABSTRACT

Microbial infections are most often countered by inflammatory responses that are initiated through the recognition of conserved microbial products by innate immune receptors and result in pathogen expulsion1-6. However, inflammation can also lead to pathology. Tissues such as the intestinal epithelium, which are exposed to microbial products, are therefore subject to stringent negative regulatory mechanisms to prevent signalling through innate immune receptors6-11. This presents a challenge to the enteric pathogen Salmonella Typhimurium, which requires intestinal inflammation to compete against the resident microbiota and to acquire the nutrients and electron acceptors that sustain its replication12,13. We show here that S. Typhimurium stimulates pro-inflammatory signalling by a unique mechanism initiated by effector proteins that are delivered by its type III protein secretion system. These effectors activate Cdc42 and the p21-activated kinase 1 (PAK1) leading to the recruitment of TNF receptor-associated factor 6 (TRAF6) and mitogen-activated protein kinase kinase kinase 7 (TAK1), and the stimulation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) inflammatory signalling. The removal of Cdc42, PAK1, TRAF6 or TAK1 prevented S. Typhimurium from stimulating NF-κB signalling in cultured cells. In addition, oral administration of a highly specific PAK inhibitor blocked Salmonella-induced intestinal inflammation and bacterial replication in the mouse intestine, although it resulted in a significant increase in the bacterial loads in systemic tissues. Thus, S. Typhimurium stimulates inflammatory signalling in the intestinal tract by engaging critical downstream signalling components of innate immune receptors. These findings illustrate the unique balance that emerges from host-pathogen co-evolution, in that pathogen-initiated responses that help pathogen replication are also important to prevent pathogen spread to deeper tissues.


Subject(s)
Host-Pathogen Interactions/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology , p21-Activated Kinases/metabolism , Animals , Bacterial Load , Cells, Cultured , Humans , Immunity, Innate , Intestines/immunology , Intestines/microbiology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , NF-kappa B/metabolism , Phosphorylation , Salmonella Infections/microbiology , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Type III Secretion Systems/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/genetics
6.
PLoS Pathog ; 12(4): e1005552, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27058235

ABSTRACT

Salmonella Typhimurium stimulates inflammatory responses in the intestinal epithelium, which are essential for its ability to replicate within the intestinal tract. Stimulation of these responses is strictly dependent on the activity of a type III secretion system encoded within its pathogenicity island 1, which through the delivery of effector proteins, triggers signaling pathways leading to inflammation. One of these effectors is SopA, a HECT-type E3 ligase, which is required for the efficient stimulation of inflammation in an animal model of Salmonella Typhimurium infection. We show here that SopA contributes to the stimulation of innate immune responses by targeting two host E3 ubiquitin ligases, TRIM56 and TRIM65. We also found that TRIM65 interacts with the innate immune receptor MDA5 enhancing its ability to stimulate interferon-ß signaling. Therefore, by targeting TRIM56 and TRIM65, SopA can stimulate signaling through two innate immune receptors, RIG-I and MDA5. These findings describe a Salmonella mechanism to modulate inflammatory responses by directly targeting innate immune signaling mechanisms.


Subject(s)
Bacterial Proteins/immunology , Host-Parasite Interactions/immunology , Immunity, Innate/immunology , Salmonella Infections/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Cell Line , Disease Models, Animal , Gene Knockout Techniques , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Real-Time Polymerase Chain Reaction , Salmonella typhimurium/immunology , Signal Transduction/immunology
7.
PLoS Pathog ; 12(3): e1005484, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26933955

ABSTRACT

Microbial infections usually lead to host innate immune responses and inflammation. These responses most often limit pathogen replication although they can also result in host-tissue damage. The enteropathogenic bacteria Salmonella Typhimurium utilizes a type III secretion system to induce intestinal inflammation by delivering specific effector proteins that stimulate signal transduction pathways resulting in the production of pro-inflammatory cytokines. We show here that a family of related Salmonella Typhimurium effector proteins PipA, GogA and GtgA redundantly target components of the NF-κB signaling pathway to inhibit transcriptional responses leading to inflammation. We show that these effector proteins are proteases that cleave both the RelA (p65) and RelB transcription factors but do not target p100 (NF-κB2) or p105 (NF-κB1). A Salmonella Typhimurium strain lacking these effectors showed increased ability to stimulate NF-κB and increased virulence in an animal model of infection. These results indicate that bacterial pathogens can evolve determinants to preserve host homeostasis and that those determinants can reduce the pathogen's virulence.


Subject(s)
Bacterial Proteins/immunology , NF-kappa B/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology , Animals , Female , Homeostasis , Humans , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Type III Secretion Systems , Virulence
8.
J Immunol ; 194(10): 4901-13, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25876760

ABSTRACT

The adenylate cyclase toxin-hemolysin (CyaA) plays a key role in the virulence of Bordetella pertussis. CyaA penetrates complement receptor 3-expressing phagocytes and catalyzes uncontrolled conversion of cytosolic ATP to the key second messenger molecule cAMP. This paralyzes the capacity of neutrophils and macrophages to kill bacteria by complement-dependent oxidative burst and opsonophagocytic mechanisms. We show that cAMP signaling through the protein kinase A (PKA) pathway activates Src homology domain 2 containing protein tyrosine phosphatase (SHP) 1 and suppresses production of bactericidal NO in macrophage cells. Selective activation of PKA by the cell-permeable analog N(6)-benzoyladenosine-3',5'-cyclic monophosphate interfered with LPS-induced inducible NO synthase (iNOS) expression in RAW264.7 macrophages, whereas inhibition of PKA by H-89 largely restored the production of iNOS in CyaA-treated murine macrophages. CyaA/cAMP signaling induced SHP phosphatase-dependent dephosphorylation of the c-Fos subunit of the transcription factor AP-1 and thereby inhibited TLR4-triggered induction of iNOS gene expression. Selective small interfering RNA knockdown of SHP-1, but not of the SHP-2 phosphatase, rescued production of TLR-inducible NO in toxin-treated cells. Finally, inhibition of SHP phosphatase activity by NSC87877 abrogated B. pertussis survival inside murine macrophages. These results reveal that an as yet unknown cAMP-activated signaling pathway controls SHP-1 phosphatase activity and may regulate numerous receptor signaling pathways in leukocytes. Hijacking of SHP-1 by CyaA action then enables B. pertussis to evade NO-mediated killing in sentinel cells of innate immunity.


Subject(s)
Adenylate Cyclase Toxin/immunology , Bordetella Infections/immunology , Macrophages/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Animals , Bordetella Infections/enzymology , Bordetella pertussis/immunology , Cell Line , Cyclic AMP , Enzyme Activation/immunology , Humans , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology
9.
PLoS One ; 9(8): e104064, 2014.
Article in English | MEDLINE | ID: mdl-25084094

ABSTRACT

Adenylate cyclase toxin (CyaA) is a key virulence factor of the whooping cough agent Bordetella pertussis. The toxin targets CD11b-expressing phagocytes and delivers into their cytosol an adenylyl cyclase (AC) enzyme that subverts cellular signaling by increasing cAMP levels. In the present study, we analyzed the modulatory effects of CyaA on adhesive, migratory and antigen presenting properties of Toll-like receptor (TLR)-activated murine and human dendritic cells (DCs). cAMP signaling of CyaA enhanced TLR-induced dissolution of cell adhesive contacts and migration of DCs towards the lymph node-homing chemokines CCL19 and CCL21 in vitro. Moreover, we examined in detail the capacity of toxin-treated DCs to induce CD4(+) and CD8(+) T cell responses. Exposure to CyaA decreased the capacity of LPS-stimulated DCs to present soluble protein antigen to CD4+ T cells independently of modulation of co-stimulatory molecules and cytokine production, and enhanced their capacity to promote CD4(+)CD25(+)Foxp3(+) T regulatory cells in vitro. In addition, CyaA decreased the capacity of LPS-stimulated DCs to induce CD8(+) T cell proliferation and limited the induction of IFN-γ producing CD8(+) T cells while enhancing IL-10 and IL-17-production. These results indicate that through activation of cAMP signaling, the CyaA may be mobilizing DCs impaired in T cell stimulatory capacity and arrival of such DCs into draining lymph nodes may than contribute to delay and subversion of host immune responses during B. pertussis infection.


Subject(s)
Adenylate Cyclase Toxin/pharmacology , Bordetella pertussis/chemistry , Cell Movement/drug effects , Dendritic Cells/cytology , Dendritic Cells/immunology , Lymphocyte Activation/drug effects , Toll-Like Receptors/metabolism , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Humans , Mice, Inbred C57BL , Solubility , T-Lymphocytes, Regulatory/drug effects
10.
PLoS One ; 8(4): e62426, 2013.
Article in English | MEDLINE | ID: mdl-23658628

ABSTRACT

Celiac disease (CD) is a gluten-responsive, chronic inflammatory enteropathy. IL-1 cytokine family members IL-1ß and IL-18 have been associated with the inflammatory conditions in CD patients. However, the mechanisms of IL-1 molecule activation in CD have not yet been elucidated. We show in this study that peripheral blood mononuclear cells (PBMC) and monocytes from celiac patients responded to pepsin digest of wheat gliadin fraction (PDWGF) by a robust secretion of IL-1ß and IL-1α and a slightly elevated production of IL-18. The analysis of the upstream mechanisms underlying PDWGF-induced IL-1ß production in celiac PBMC show that PDWGF-induced de novo pro-IL-1ß synthesis, followed by a caspase-1 dependent processing and the secretion of mature IL-1ß. This was promoted by K+ efflux and oxidative stress, and was independent of P2X7 receptor signaling. The PDWGF-induced IL-1ß release was dependent on Nod-like receptor family containing pyrin domain 3 (NLRP3) and apoptosis-associated speck like protein (ASC) as shown by stimulation of bone marrow derived dendritic cells (BMDC) from NLRP3(-/-) and ASC(-/-) knockout mice. Moreover, treatment of human PBMC as well as MyD88(-/-) and Toll-interleukin-1 receptor domain-containing adaptor-inducing interferon-ß (TRIF)(-/-) BMDC illustrated that prior to the activation of caspase-1, the PDWGF-triggered signal constitutes the activation of the MyD88/TRIF/MAPK/NF-κB pathway. Moreover, our results indicate that the combined action of TLR2 and TLR4 may be required for optimal induction of IL-1ß in response to PDWGF. Thus, innate immune pathways, such as TLR2/4/MyD88/TRIF/MAPK/NF-κB and an NLRP3 inflammasome activation are involved in wheat proteins signaling and may play an important role in the pathogenesis of CD.


Subject(s)
Carrier Proteins/immunology , Gliadin/immunology , Inflammasomes/drug effects , Interleukin-1beta/immunology , Leukocytes, Mononuclear/drug effects , Peptide Fragments/pharmacology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adult , Animals , Carrier Proteins/genetics , Celiac Disease , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Gliadin/chemistry , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1beta/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Male , Mice , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Pepsin A , Primary Cell Culture , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
11.
PLoS Pathog ; 8(4): e1002580, 2012.
Article in English | MEDLINE | ID: mdl-22496638

ABSTRACT

Bordetella adenylate cyclase toxin-hemolysin (CyaA) penetrates the cytoplasmic membrane of phagocytes and employs two distinct conformers to exert its multiple activities. One conformer forms cation-selective pores that permeabilize phagocyte membrane for efflux of cytosolic potassium. The other conformer conducts extracellular calcium ions across cytoplasmic membrane of cells, relocates into lipid rafts, translocates the adenylate cyclase enzyme (AC) domain into cells and converts cytosolic ATP to cAMP. We show that the calcium-conducting activity of CyaA controls the path and kinetics of endocytic removal of toxin pores from phagocyte membrane. The enzymatically inactive but calcium-conducting CyaA-AC⁻ toxoid was endocytosed via a clathrin-dependent pathway. In contrast, a doubly mutated (E570K+E581P) toxoid, unable to conduct Ca²âº into cells, was rapidly internalized by membrane macropinocytosis, unless rescued by Ca²âº influx promoted in trans by ionomycin or intact toxoid. Moreover, a fully pore-forming CyaA-ΔAC hemolysin failed to permeabilize phagocytes, unless endocytic removal of its pores from cell membrane was decelerated through Ca²âº influx promoted by molecules locked in a Ca²âº-conducting conformation by the 3D1 antibody. Inhibition of endocytosis also enabled the native B. pertussis-produced CyaA to induce lysis of J774A.1 macrophages at concentrations starting from 100 ng/ml. Hence, by mediating calcium influx into cells, the translocating conformer of CyaA controls the removal of bystander toxin pores from phagocyte membrane. This triggers a positive feedback loop of exacerbated cell permeabilization, where the efflux of cellular potassium yields further decreased toxin pore removal from cell membrane and this further enhances cell permeabilization and potassium efflux.


Subject(s)
Adenylate Cyclase Toxin/pharmacology , Cell Membrane Permeability/drug effects , Macrophages/metabolism , Membrane Microdomains/metabolism , Potassium/metabolism , Animals , Cell Line , Clathrin/metabolism , Endocytosis/drug effects , Ion Transport/drug effects , Macrophages/cytology , Mice
12.
Infect Immun ; 80(3): 1181-92, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22215742

ABSTRACT

The Bordetella adenylate cyclase toxin-hemolysin (CyaA; also called ACT or AC-Hly) targets CD11b-expressing phagocytes and translocates into their cytosol an adenylyl cyclase (AC) that hijacks cellular signaling by conversion of ATP to cyclic AMP (cAMP). Intriguingly, insertion of large passenger peptides removes the enzymatic activity but not the cell-invasive capacity of the AC domain. This has repeatedly been exploited for delivery of heterologous antigens into the cytosolic pathway of CD11b-expressing dendritic cells by CyaA/AC(-) toxoids, thus enabling their processing and presentation on major histocompatibility complex (MHC) class I molecules to cytotoxic CD8(+) T lymphocytes (CTLs). We produced a set of toxoids with overlapping deletions within the first 371 residues of CyaA and showed that the structure of the AC enzyme does not contain any sequences indispensable for its translocation across target cell membrane. Moreover, replacement of the AC domain (residues 1 to 371) with heterologous polypeptides of 40, 146, or 203 residues yielded CyaAΔAC constructs that delivered passenger CTL epitopes into antigen-presenting cells (APCs) and induced strong antigen-specific CD8(+) CTL responses in vivo in mice and ex vivo in human peripheral blood mononuclear cell cultures. This shows that the RTX (repeats in toxin) hemolysin moiety, consisting of residues 374 to 1706 of CyaA, harbors all structural information involved in translocation of the N-terminal AC domain across target cell membranes. These results decipher the extraordinary capacity of the AC domain of CyaA to transport large heterologous cargo polypeptides into the cytosol of CD11b(+) target cells and pave the way for the construction of CyaAΔAC-based polyvalent immunotherapeutic T cell vaccines.


Subject(s)
Adenylate Cyclase Toxin/metabolism , Antigen-Presenting Cells/metabolism , Cell Membrane/metabolism , Dendritic Cells/metabolism , Adenylate Cyclase Toxin/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Protein Transport , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Toxoids/genetics , Toxoids/metabolism
13.
J Cell Mol Med ; 15(4): 938-48, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20406323

ABSTRACT

In genetically predisposed individuals, ingestion of wheat gliadin provokes a T-cell-mediated enteropathy, celiac disease. Gliadin fragments were previously reported to induce phenotypic maturation and Th1 cytokine production by human dendritic cells (DCs) and to boost their capacity to stimulate allogeneic T cells. Here, we monitor the effects of gliadin on migratory capacities of DCs. Using transwell assays, we show that gliadin peptic digest stimulates migration of human DCs and their chemotactic responsiveness to the lymph node-homing chemokines CCL19 and CCL21. The gliadin-induced migration is accompanied by extensive alterations of the cytoskeletal organization, with dissolution of adhesion structures, podosomes, as well as up-regulation of the CC chemokine receptor (CCR) 7 on cell surface and induction of cyclooxygenase (COX)-2 enzyme that mediates prostaglandin E2 (PGE2) production. Blocking experiments confirmed that gliadin-induced migration is independent of the TLR4 signalling. Moreover, we showed that the α-gliadin-derived 31-43 peptide is an active migration-inducing component of the digest. The migration promoted by gliadin fragments or the 31-43 peptide required activation of p38 mitogen-activated protein kinase (MAPK). As revealed using p38 MAPK inhibitor SB203580, this was responsible for DC cytoskeletal transition, CCR7 up-regulation and PGE2 production in particular. Taken together, this study provides a new insight into pathogenic features of gliadin fragments by demonstrating their ability to promote DC migration, which is a prerequisite for efficient priming of naive T cells, contributing to celiac disease pathology.


Subject(s)
Chemotaxis/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gliadin/pharmacology , Peptide Fragments/pharmacology , Chemokine CCL19/pharmacology , Chemokine CCL21/pharmacology , Cyclooxygenase 2/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dendritic Cells/enzymology , Dinoprostone/biosynthesis , Enzyme Activation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Models, Biological , Receptors, CCR7/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
14.
FEMS Microbiol Rev ; 34(6): 1076-112, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20528947

ABSTRACT

Repeats-in-toxin (RTX) exoproteins of Gram-negative bacteria form a steadily growing family of proteins with diverse biological functions. Their common feature is the unique mode of export across the bacterial envelope via the type I secretion system and the characteristic, typically nonapeptide, glycine- and aspartate-rich repeats binding Ca(2+) ions. In this review, we summarize the current state of knowledge on the organization of rtx loci and on the biological and biochemical activities of therein encoded proteins. Applying several types of bioinformatic screens on the steadily growing set of sequenced bacterial genomes, over 1000 RTX family members were detected, with the biological functions of most of them remaining to be characterized. Activities of the so far characterized RTX family members are then discussed and classified according to functional categories, ranging from the historically first characterized pore-forming RTX leukotoxins, through the large multifunctional enzymatic toxins, bacteriocins, nodulation proteins, surface layer proteins, up to secreted hydrolytic enzymes exhibiting metalloprotease or lipase activities of industrial interest.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Gram-Negative Bacteria/metabolism , Multigene Family , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Gene Expression Regulation, Bacterial , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/genetics , Protein Transport
15.
J Immunol ; 181(8): 5587-97, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832717

ABSTRACT

Adenylate cyclase toxin (CyaA or ACT) is a key virulence factor of pathogenic Bordetellae. It penetrates phagocytes expressing the alpha(M)beta(2) integrin (CD11b/CD18, Mac-1 or CR3) and paralyzes their bactericidal capacities by uncontrolled conversion of ATP into a key signaling molecule, cAMP. Using pull-down activity assays and transfections with mutant Rho family GTPases, we show that cAMP signaling of CyaA causes transient and selective inactivation of RhoA in mouse macrophages in the absence of detectable activation of Rac1, Rac2, or RhoG. This CyaA/cAMP-induced drop of RhoA activity yielded dephosphorylation of the actin filament severing protein cofilin and massive actin cytoskeleton rearrangements, which were paralleled by rapidly manifested macrophage ruffling and a rapid and unexpected loss of macropinocytic fluid phase uptake. As shown in this study for the first time, CyaA/cAMP signaling further caused a rapid and near-complete block of complement-mediated phagocytosis. Induction of unproductive membrane ruffling, hence, represents a novel sophisticated mechanism of down-modulation of bactericidal activities of macrophages and a new paradigm for action of bacterial toxins that hijack host cell signaling by manipulating cellular cAMP levels.


Subject(s)
Adenylate Cyclase Toxin/immunology , Bordetella pertussis/immunology , Macrophage-1 Antigen/immunology , Macrophages/immunology , Signal Transduction/immunology , Whooping Cough/immunology , rho GTP-Binding Proteins/immunology , Actin Cytoskeleton/immunology , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/immunology , Actin Depolymerizing Factors/metabolism , Adenylate Cyclase Toxin/metabolism , Animals , Bordetella pertussis/enzymology , CD11b Antigen/genetics , CD11b Antigen/immunology , CD18 Antigens/genetics , CD18 Antigens/immunology , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Cyclic AMP/immunology , Female , GTP Phosphohydrolases/immunology , GTP Phosphohydrolases/metabolism , Macrophage-1 Antigen/metabolism , Macrophages/metabolism , Mice , Neuropeptides/immunology , Neuropeptides/metabolism , Whooping Cough/enzymology , rac GTP-Binding Proteins/immunology , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein , RAC2 GTP-Binding Protein
16.
Vaccine ; 26(47): 5935-43, 2008 Nov 05.
Article in English | MEDLINE | ID: mdl-18804138

ABSTRACT

Sterile immunity against malaria can be achieved by the induction of IFNgamma-producing CD8(+) T cells that target infected hepatocytes presenting epitopes of the circumsporozoite protein (CSP). In the present study we evaluate the protective efficacy of a heterologous prime/boost immunization protocol based on the delivery of the CD8(+) epitope of Plasmodium berghei CSP into the MHC class I presentation pathway, by either a type III secretion system of live recombinant Salmonella and/or by direct translocation of a recombinant Bordetella adenylate cyclase toxoid fusion (ACT-CSP) into the cytosol of professional antigen-presenting cells (APCs). A single intraperitoneal application of the recombinant ACT-CSP toxoid, as well as a single oral immunization with the Salmonella vaccine, induced a specific CD8(+) T cell response, which however conferred only a partial protection on mice against a subsequent sporozoite challenge. In contrast, a heterologous prime/boost vaccination with the live Salmonella followed by ACT-CSP led to a significant enhancement of the CSP-specific T cell response and induced complete protection in all vaccinated mice.


Subject(s)
Bordetella , Malaria/prevention & control , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Recombinant Fusion Proteins/immunology , Salmonella typhimurium , Adenylate Cyclase Toxin/genetics , Adenylate Cyclase Toxin/immunology , Adenylate Cyclase Toxin/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bordetella/genetics , Bordetella/immunology , Bordetella/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunization , Immunization, Secondary , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism
17.
Infect Immun ; 74(4): 2277-85, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16552058

ABSTRACT

The adenylate cyclase toxoid (ACT) of Bordetella pertussis is capable of delivering its N-terminal catalytic domain into the cytosol of CD11b-expressing professional antigen-presenting cells such as myeloid dendritic cells. This allows delivery of CD8+ T-cell epitopes to the major histocompatibility complex (MHC) class I presentation pathway. Recombinant detoxified ACT containing an epitope of the Plasmodium berghei circumsporozoite protein (CSP), indeed, induced a specific CD8+ T-cell response in immunized mice after a single application, as detected by MHC multimer staining and gamma interferon (IFN-gamma) ELISPOT assay. This CSP-specific response could be significantly enhanced by prime-boost immunization with recombinant ACT in combination with anti-CTLA-4 during the boost immunization. This increased response was accompanied by complete protection in a number of mice after a challenge with P. berghei sporozoites. Transient blockade of CTLA-4 may overcome negative regulation and hence provide a strategy to enhance the efficacy of a vaccine by amplifying the number of responding T cells.


Subject(s)
Adenylate Cyclase Toxin/immunology , Antigens, Differentiation/immunology , Bordetella pertussis/immunology , Liver Diseases, Parasitic/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenylate Cyclase Toxin/genetics , Animals , Antigens, CD , Antigens, Differentiation/metabolism , Bordetella pertussis/enzymology , Bordetella pertussis/genetics , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Cells, Cultured , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class I/immunology , Immunization, Secondary , Liver Diseases, Parasitic/parasitology , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology , Plasmodium berghei/genetics , Protozoan Proteins , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/metabolism
18.
Curr Opin Microbiol ; 9(1): 69-75, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16406775

ABSTRACT

Bordetella that infect mammals produce a multifunctional repeat in toxin (RTX) adenylate cyclase toxin known as CyaA, an excellent example of bacterial sophistication in subverting host defense. Recent reports show that interaction of CyaA with tracheal epithelial cells aids adhesion of Bordetella to ciliated mucosa and induces production of the pro-inflammatory cytokine interleukin, IL-6. Myeloid phagocytes, attracted to the site of infection are the target of freshly secreted CyaA that binds to the alpha(M)beta2 integrin (CD11b/CD18), penetrates cells and promptly suppresses their bactericidal functions by converting cellular ATP to cAMP. Such uncontrolled cAMP signaling can also drive CD11b-expressing immature dendritic cells into a semi-mature state, possibly hijacking them to shape the local adaptive immune response towards tolerance of the pathogen.


Subject(s)
Adenylate Cyclase Toxin/toxicity , Bordetella Infections/microbiology , Bordetella/immunology , Bordetella/pathogenicity , Respiratory Tract Infections/microbiology , Adenylate Cyclase Toxin/metabolism , Animals , Bacterial Adhesion , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Bordetella Infections/immunology , CD11b Antigen/metabolism , CD18 Antigens/metabolism , Dendritic Cells/immunology , Epithelial Cells/microbiology , Humans , Interleukin-6/biosynthesis , Phagocytes/immunology , Phagocytes/microbiology , Respiratory Mucosa/microbiology , Respiratory Tract Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...