Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About IAI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Infection and Immunity
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • Archive
    • Minireviews
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About IAI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Microbial Immunity and Vaccines

Alternative Endogenous Protein Processing via an Autophagy-Dependent Pathway Compensates for Yersinia-Mediated Inhibition of Endosomal Major Histocompatibility Complex Class II Antigen Presentation

Holger Rüssmann, Klaus Panthel, Brigitte Köhn, Stefan Jellbauer, Sebastian E. Winter, Sara Garbom, Hans Wolf-Watz, Sigrid Hoffmann, Silke Grauling-Halama, Gernot Geginat
Holger Rüssmann
1Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Ludwig-Maximilians-Universität München, 80336 Munich
2Institut für Mikrobiologie, Immunologie und Laboratoriumsmedizin, HELIOS Klinikum Emil von Behring, 14165 Berlin, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: holger.ruessmann@helios-kliniken.de
Klaus Panthel
1Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Ludwig-Maximilians-Universität München, 80336 Munich
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Brigitte Köhn
1Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Ludwig-Maximilians-Universität München, 80336 Munich
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stefan Jellbauer
1Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Ludwig-Maximilians-Universität München, 80336 Munich
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sebastian E. Winter
1Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Ludwig-Maximilians-Universität München, 80336 Munich
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sara Garbom
3Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hans Wolf-Watz
3Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sigrid Hoffmann
4Zentrum für Medizinische Forschung, Fakultät für Medizin Mannheim der Universität Heidelberg, 68167 Mannheim
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Silke Grauling-Halama
4Zentrum für Medizinische Forschung, Fakultät für Medizin Mannheim der Universität Heidelberg, 68167 Mannheim
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gernot Geginat
5Institut für Medizinische Mikrobiologie und Hygiene, Fakultät für Medizin Mannheim der Universität Heidelberg, 68167 Mannheim, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/IAI.00155-10
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Extracellular Yersinia pseudotuberculosis employs a type III secretion system (T3SS) for translocating virulence factors (Yersinia outer proteins [Yops]) directly into the cytosol of eukaryotic cells. Recently, we used YopE as a carrier molecule for T3SS-dependent secretion and translocation of listeriolysin O (LLO) from Listeria monocytogenes. We demonstrated that translocation of chimeric YopE/LLO into the cytosol of macrophages by Yersinia results in the induction of a codominant antigen-specific CD4 and CD8 T-cell response in orally immunized mice. In this study, we addressed the requirements for processing and major histocompatibility complex (MHC) class II presentation of chimeric YopE proteins translocated into the cytosol of macrophages by the Yersinia T3SS. Our data demonstrate the ability of Yersinia to counteract exogenous MHC class II antigen presentation of secreted hybrid YopE by the action of wild-type YopE and YopH. In the absence of exogenous MHC class II antigen presentation, an alternative pathway was identified for YopE fusion proteins originating in the cytosol. This endogenous antigen-processing pathway was sensitive to inhibitors of phagolysosomal acidification and macroautophagy, but it did not require the function either of the proteasome or of transporters associated with antigen processing. Thus, by an autophagy-dependent mechanism, macrophages are able to compensate for the YopE/YopH-mediated inhibition of the endosomal MHC class II antigen presentation pathway for exogenous antigens. This is the first report demonstrating that autophagy might enable the host to mount an MHC class II-restricted CD4 T-cell response against translocated bacterial virulence factors. We provide critical new insights into the interaction between the mammalian immune system and a human pathogen.

Protein antigens are recognized by T cells as short peptide fragments bound either to major histocompatibility class (MHC) I or to MHC class II molecules on the surface of antigen-presenting cells (APCs). The location of antigens in distinct intracellular compartments of APCs influences their proteolytic processing as well as access to MHC molecules (16, 39). Classically, peptides generated in the cytosol (e.g., derived from viral proteins) by proteasomal degradation are bound to MHC class I molecules after transport across the endoplasmic reticulum membrane by the transporters associated with antigen processing (TAP) (90). Subsequently, MHC class I molecules present these endogenous antigenic peptides to CD8 T cells. In contrast, exogenous antigens (e.g., antigens derived from engulfed bacteria and soluble antigens) are directed into the endosomal/lysosomal pathway for degradation (63). In late endosomal compartments, degraded protein fragments interact with MHC class II molecules and are further trimmed into peptides for presentation to CD4 T cells.

However, the synchrony of this system has been challenged by biochemical and functional studies of professional and nonprofessional APCs. Epitopes derived from a diverse pool of cytosolic antigens such as metabolic enzymes, cytoskeletal proteins, and viral and tumor antigens have been identified to be presented in the context of both murine and human MHC class II molecules (20, 51, 52, 53, 71, 88). Several different alternative pathways for delivering antigens into the MHC class II pathway have been described. Lich et al. reported that cytoplasmic processing by the proteasome and calpain is required for efficient processing of the autoantigen glutamate decarboxylase to CD4 T cells (51). Also, a proteasome/TAP-dependent pathway was shown to be important for the presentation of MHC class II peptides from the influenza virus (83). In contrast, other studies using human B lymphoblastoid cells revealed that TAP is not involved in the transport of cytosolic peptides to MHC class II molecules (52, 53). Alternatively, autophagy in the form of either macroautophagy (27, 30, 56, 60) or chaperone-mediated autophagy (19, 23, 24, 60, 91) results in the transport of cytosolic peptides and proteins directly into endosomes and lysosomes. Two essential components of the chaperone-mediated autophagy pathway are Lamp-2a and the heat shock cognate protein hsc70. The latter molecule is an accessory chaperone that intersects target proteins in the cytoplasm and facilitates their delivery to Lamp-2a, a lysosomal membrane protein.

Pathogenic yersiniae are Gram-negative bacteria that cause a wide range of diseases in humans, ranging from bubonic plague, caused by Yersinia pestis, to self-limiting gastroenteritis and lymphadenitis, caused by the enteric pathogens Yersinia pseudotuberculosis and Yersinia enterocolitica. Results from in vitro experiments and the mouse infection model revealed that the last two bacterial species have acquired a complex arsenal of effector proteins to overcome host defense mechanisms. These major pathogenicity factors are located on a 70-kb virulence plasmid, which encodes a protein export apparatus called the type III secretion system (T3SS) (21, 35). T3SS is a complex bacterial organelle that provides Gram-negative pathogens with a unique virulence mechanism enabling them to translocate bacterial effector proteins directly into the host cell cytosol. At least six effector proteins, Yersinia outer proteins (Yops), namely, YopE, YopH, YopM, YopO/YpkA, YopP/YopJ, and YopT, are injected into the cytosol of eukaryotic cells in a T3SS-dependent manner (22). The main function of these Yops is to inhibit the immune response of the host. Four Yops (YopE, YopH, YopO/YpkA, and YopT) are involved in inhibiting phagocytosis of yersiniae by disrupting the cytoskeleton of polymorphonuclear leukocytes and macrophages (12, 33, 40, 68). Thus, the consequence of this translocation process is that pathogenic yersiniae survive and proliferate at extracellular sites in the infected host (77).

Our laboratory has described and analyzed in detail the potential of YopE to be a carrier molecule for heterologous antigen delivery by Yersinia (74). YopE is a translocated GTPase-activating molecule that can downregulate Rho activity, leading to actin filament disruption and inhibition of phagocytosis by macrophages (10, 69, 87). The N-terminal 18 amino acids (aa) of YopE fused to a large protein fragment of the p60 antigen from the intracellular pathogen Listeria monocytogenes were sufficient for T3SS-dependent secretion to the extracellular environment of Yersinia-infected target cells (74). In contrast, fusion of p60 to the N-terminal 138 aa of YopE resulted in translocation of the chimeric protein into the cytosol of host cells. As expected, T-cell activation assays revealed that the cytosolic delivery of this hybrid protein was a prerequisite to induce a p60-specific MHC class I-restricted CD8 T-cell response (74). Because translocated p60 easily enters the endogenous cytosolic antigen presentation pathway, it is processed like an endogenously synthesized antigen. In a more recent publication, we used Y. pseudotuberculosis expressing YopE fused to listeriolysin O (LLO) from Listeria to assess the influence of secreted versus translocated antigen display on in vitro antigen presentation and in vivo T-cell priming in the oral mouse infection model (72). We constructed two different plasmid-encoded hybrid YopE/LLO proteins. By engaging the above-mentioned well-defined secretion and translocation domains of YopE (79) fused to aa 51 to 363 of LLO, chimeric YopE was expressed either in secreted or in translocated form (72). Biochemical fractionation of Yersinia-infected macrophage-like P338D1 cells clearly revealed that YopE from aa 1 to 38 (YopE1-138)/LLO was translocated to the cytosol of host cells, whereas YopE1-18/LLO lacking the translocation domain was efficiently secreted to the culture medium but was not detected in the P338D1 cell lysate containing cytosolic proteins. Thus, it was theoretically expected that the well-secreted version of chimeric YopE/LLO could have the potential to enter the exogenous MHC class II antigen presentation pathway for proper CD4 T-cell priming. Strikingly, results from in vitro antigen presentation assays and also the enumeration of LLO-specific CD4 T cells from infected mice indicated a superior efficacy of translocated over secreted LLO for MHC class II antigen presentation and CD4 T-cell induction, respectively (72).

This study addresses the requirements for processing and MHC class II presentation of chimeric YopE proteins translocated into the cytosol of macrophages by the T3SS of Yersinia pseudotuberculosis. Our data demonstrate the ability of Yersinia to counteract exogenous MHC class II antigen presentation of secreted hybrid YopE by the action of wild-type YopE and YopH. However, a distinct MHC class II antigen presentation pathway was identified for YopE fusion proteins originating from the cytosol. Presentation of cytoplasm-derived chimeric YopE requires acidification of the phagolysosome and is sensitive to inhibitors of autophagy.

MATERIALS AND METHODS

Plasmids, bacterial strains, and growth conditions.The plasmids (pACYC184 derivatives) and Y. pseudotuberculosis strains used in this study are listed in Table 1. Translational fusions between different lengths of YopE or YopH with LLO or p60 were constructed by standard PCR cloning procedures, as described previously (72, 73). All resulting protein fusions were checked by DNA sequencing. Escherichia coli χ6060 was used as an intermediate host for cloning experiments. The chimeric proteins used in this study are tagged at their C termini with an M45 epitope tag (MDRSRDRLPPFETETRIL) that is derived from the E4-6/7 protein of adenovirus (58). Overnight cultures of Y. pseudotuberculosis strains were grown in Luria-Bertani (LB) medium at 27°C. On the next day, cultures were diluted and incubated at 37°C for 4 h to allow expression of components and targets of the T3SS encoded by the Yersinia virulence plasmid (74). When required, the antibiotics kanamycin (30 μg/ml) and chloramphenicol (20 μg/ml) were added.

View this table:
  • View inline
  • View popup
TABLE 1.

Strains and plasmids used in this study

Detection of secreted hybrid YopE and YopH proteins by Western blot analysis.Secreted proteins from Yersinia culture supernatants were prepared as follows. Briefly, bacterial supernatants were passed through a 0.45-μm-pore-size syringe filter to remove bacteria. Protein in the bacterium-free medium was precipitated by the addition of cold trichloroacetic acid to 10% (vol/vol) and incubated for 2 h on ice. The protein was collected by centrifugation at 10,000 × g and 4°C for 20 min. The pellets were washed in 0.8 ml cold acetone, dried, and resuspended in phosphate-buffered saline (PBS) buffered with 80 mM Tris-HCl, pH 8. Samples corresponding to 500 μl culture supernatant were separated in a 10% discontinuous sodium dodecyl sulfate (SDS)-polyacrylamide gel and transferred to nitrocellulose membranes. Hybrid proteins were detected by immunoblot analysis. Western blots were treated with a monoclonal antibody against M45, followed by incubation with a horseradish peroxidase-labeled antimouse antibody. Blots were developed by using a chemiluminescence kit.

Measurement of antigen uptake by immunofluorescence analysis.Macrophage-like P388D1 cells were grown on glass coverslips to 60% confluence. One hour before the addition of bacteria, Dulbecco's modified Eagle's medium (DMEM) was replaced by 500 μl of Hank's balanced salt solution (HBSS). P388D1 cells were infected with Y. pseudotuberculosis strain pIB102 or pIB251 at a multiplicity of infection (MOI) of 5 to 10 for 15 min. Then, macrophage-like cells were incubated with 50 μg/ml dye-quenched (DQ) ovalbumin (OVA)-fluorescein isothiocyanate (FITC) (Molecular Probes, Eugene, OR) for 30 min at 37°C, washed three times with HBSS to remove non-cell-associated bacteria and DQ-OVA-FITC, and fixed in PBS-3.7% formaldehyde. Extracellular yersiniae were stained with an anti-Y. pseudotuberculosis lipopolysaccharide (LPS) polyclonal rabbit antiserum (a kind gift of R. Rosqvist, Umeå University, Umeå, Sweden) and a secondary anti-rabbit tetramethylrhodamine isothiocyanate (TRITC) conjugate (1:100 in PBS-3% bovine serum albumin; Sigma, St. Louis, MO). After permeabilization of P388D1 cells (3 min in PBS-0.1% Triton X-100), extra- and intracellular yersiniae were stained with a polyclonal anti-Y. pseudotuberculosis LPS antiserum and a secondary anti-rabbit 7-amino-4-methylcoumarin-3-acetic acid (AMCA) conjugate (1:100 in PBS-3% bovine serum albumin; Jackson Immuno Research, Newmarket, United Kingdom). Coverslips were mounted on glass slides and analyzed by fluorescence microscopy. Experiments were repeated at least three times.

T-cell lines.CD8 T-cell lines specific for p60217-225 and LLO91-99 and CD4 T cells against the CD4 T-cell epitope p60301-312 were derived from spleens of L. monocytogenes-infected BALB/c mice. CD4 T-cell lines specific for the H-2b-restricted CD4 T-cell epitopes p60177-188 and LLO190-201 were established from spleens 14 days after intravenous infection of C57BL/6 mice with 1 × 103 CFU L. monocytogenes. All CD8 T-cell lines were propagated by repeated restimulation with P815 cells transfected with the human B7.1 gene (P815/B7) in the presence of the appropriate synthetic peptide in medium supplemented with interleukin-2 (IL-2) as described previously (38). All CD4 T-cell lines were repeatedly restimulated with syngeneic mitomycin C-inactivated splenocytes as APCs in the presence of 10−6 M peptide. The T-cell culture medium was an alpha modification of Eagle's medium supplemented with glutamine, penicillin, streptomycin, 10% fetal calf serum (FCS), 100 U/ml recombinant murine IL-2 (R&D Systems, Minneapolis, MN), and 2 × 10−5 M 2-mercaptoethanol.

BMMs.All antigen presentation studies were performed with bone marrow-derived macrophages (BMMs). Female C57BL/6 JO1aHsd (H-2b) and CB6F1 (H-2b × H-2d) mice were purchased (Centre d'Elevage R. Janvier, Le Genest-St. Isle, France), kept under conventional conditions, and used as bone marrow donors at 8 to 16 weeks of age. Macrophages were seeded at a density of 1 × 105 cells per well in 96-well flat-bottom tissue culture plates in DMEM supplemented with 10% FCS and 10 ng/ml granulocyte-macrophage colony-stimulating factor (GM-CSF; R&D Systems). BMM cultures were fed with 100 μl GM-CSF-supplemented medium every 4 days and were used after 12 to 16 days of culture. Nonadherent cells were removed by thorough washing before use.

In vitro antigen presentation studies.Direct antigen presentation assays were performed as described previously (78) with some modifications. BMMs to be used in antigen presentation assays were cultured for 24 h in medium supplemented with 10 ng/ml gamma interferon (IFN-γ; R&D Systems). Macrophages were infected with yersiniae for 4 h at the indicated MOI. Cells were fixed with 1% paraformaldehyde before addition of T cells. After overnight incubation, T-cell activation was assessed by measuring the IFN-γ concentration in culture supernatants with an IFN-γ specific enzyme-linked immunosorbent assay (ELISA) that binds and detects IFN-γ with a pair of specific monoclonal antibodies. Results were corrected for dilution of the sample to yield the sample concentration in ng/ml.

Inhibition experiments were performed in the presence of proteasome inhibitors (epoxomycin, lactacystin), an inhibitor of tripeptidyl peptidase II (TPPII; alanyl-alanyl-phenylalanine chloromethyl ketone [AAF-CMK]), an inhibitor of endosomal proteases (leupeptin), an inhibitor of lysosomal acidification (chloroquin), or inhibitors of the autophagy pathway (3-methyladenine [3-MA] and wortmannin) at the indicated concentrations. Brefeldin A was used to inhibit the export of newly folded MHC class II molecules. Inhibitors were generally added 30 to 60 min before infection of cells.

For exogenous loading experiments either purified p60, whole concentrated L. monocytogenes culture supernatant (SN), or nonconcentrated supernatant from Yersinia pseudotuberculosis was used. The bacterial murein hydrolase p60 was purified from supernatants of L. monocytogenes MR1(pGB363-1p60) cultures by preparative SDS-PAGE and subsequent gel elution, as described previously (36). Concentrated (100 ×) L. monocytogenes sv1/2a EGD supernatant was prepared as described previously (61). Yersinia pseudotuberculosis supernatant was prepared after 6 h infection (MOI, 10) of CBA/J-derived BMMs (H-2k haplotype) with pIB102(pHR429) or pIB102(pHR430). Supernatants were filtrated through a 0.22-μm-pore-size filter before use.

Western blot analysis of translocated hybrid YopE/LLO in Yersinia-infected BMMs.The detection of translocated chimeric YopE/LLO was carried out as described previously (72). Briefly, BMM monolayers were grown in 100-mm-diameter tissue culture plates in DMEM supplemented with 5% FCS. Chemical inhibitors were generally added 30 to 60 min before infection of cells. BMMs were infected with Y. pseudotuberculosis strain pIB102(pHR430) expressing translocated YopE/LLO proteins with an MOI of 10 in 2.5 ml of HBSS at 37°C. Prior to infection, bacterial overnight cultures (LB medium, 27°C) were diluted and incubated at 37°C for 4 h. After infection for 5 h, nonadherent bacteria were removed and the cells were washed with HBSS. The infection supernatant was combined with the material from the washes and centrifuged at 8,000 × g for 20 min. The pellet containing nonadherent bacteria was resuspended in 200 μl of phosphate-buffered saline (fraction of non-cell-associated bacteria). Infected BMMs were incubated for 30 min with DMEM containing 100 μg of gentamicin/ml to kill the cell-associated extracellular bacteria. The cells were then treated with 30 μg of proteinase K/ml in HBSS for 15 min at 37°C to eliminate cell surface-associated Yops. After proteinase K treatment, 3 ml of chilled HBSS containing 2 mM phenylmethylsulfonyl fluoride was added. Cells detached during the proteinase K treatment and were subsequently collected by low-speed centrifugation (600 × g for 10 min) and lysed in 1 ml of HBSS containing 0.1% Triton X-100 and 1 mM phenylmethylsulfonyl fluoride. Then, the cell lysate was centrifuged at 15,000 × g for 10 min. The supernatant was filtered through a 0.45-μm-pore-size syringe filter, and proteins were precipitated in the presence of 10% trichloroacetic acid (the fraction of Triton X-100-soluble P388D1 cell lysate containing translocated proteins).

Statistical analysis.The statistical significance of the results was analyzed using the t test at the 0.05 significance level and, if more than two experimental groups were compared, the Tukey multiple-comparison test at the 0.05 significance level (84). Calculations were performed using WINKS statistical analysis software (TexaSoft, Cedar Hill, TX).

RESULTS

MHC class II antigen presentation of exogenously loaded YopE1-18/LLO and YopE1-138/LLO by macrophages.In this study, two previously described plasmid vectors have been used (72). Plasmid pHR429 encodes the N-terminal 18 aa of YopE fused to aa 51 to 363 of LLO, resulting in a hybrid protein that contains the secretion domain but that lacks the translocation domain of YopE. Plasmid pHR430 bears the genetic information for YopE1-138/LLO51-363. This hybrid protein contains both the secretion and translocation domains of YopE. To facilitate recognition, both chimeric molecules were tagged at their C termini with an adenoviral M45 epitope (58, 72). Transcription of plasmid-borne gene fusions is mediated by the wild-type yopE promoter that is regulated by the Yersinia T3SS.

In a first set of experiments, we wanted to answer the question whether both types of hybrid YopE/LLO proteins are presented by MHC class II after exogenous loading of macrophages with these antigens. In Fig. 1A it is demonstrated that wild-type Y. pseudotuberculosis strains pIB102(pHR429) and pIB102(pHR430) secrete similar amounts of YopE1-18/LLO or YopE1-138/LLO into bacterial culture supernatants. BMMs derived from CB6F1 mice were incubated with different concentrations of these supernatants (Fig. 1B), and MHC class II antigen presentation was tested with an H-2Ab-restricted CD4 T-cell line specific for LLO190-201 (37). Loading of APCs with either chimeric YopE1-18/LLO or YopE1-138/LLO resulted in pronounced stimulation of LLO190-201-specific CD4 T cells. T-cell activation in the two experimental groups did not differ significantly.

FIG. 1.
  • Open in new tab
  • Download powerpoint
FIG. 1.

Exogenous MHC class II antigen presentation of extracellular hybrid YopE/LLO proteins. Secreted proteins from Yersinia culture supernatants were prepared as described in Materials and Methods. (A) The presence of chimeric YopE/LLO proteins was examined in supernatants from pIB102(pHR429) (lane 1) and pIB102(pHR430) (lane 2) by Western blot analysis. Hybrid YopE/LLO proteins were detected by protein immunoblotting with a monoclonal antibody directed against the M45 tag attached to their C termini. (B) MHC class II antigen presentation of hybrid YopE/LLO proteins was tested with an LLO190-201-specific CD4 T-cell line after external loading of BMMs with the indicated dilution of a Yersinia culture supernatant which was prepared as described in Materials and Methods. The concentration is indicated as MOI equivalents. T-cell activation was measured by determination of the amount of IFN-γ secreted into the culture supernatant. Means and standard deviations of triplicate determinations are indicated. The results of one out of two independent experiments are shown.

YopE and YopH inhibit exogenous antigen uptake and exogenous MHC class II antigen presentation by APCs infected with Y. pseudotuberculosis.In Y. pseudotuberculosis, three translocated T3SS proteins are known to disturb the cytoskeleton dynamics of macrophages and inhibit phagocytosis (40). YopE is a GTPase-activating protein that is active toward G proteins from the Rho family (10, 87). The tyrosine phosphatase YopH (42) disturbs focal adhesion sites by dephosphorylating the focal adhesion kinase (FAK), p130Cas, Fyn-binding protein, paxillin, and SKAP-HOM (Src Kinase-associated phosphoprotein 55 homologue) (9, 11, 45, 62). The threonine kinase YpkA is activated by actin and binds to GTP- and GDP-bound forms of Rho GTPases (31, 49).

In a next set of experiments, we investigated whether the antiphagocytic molecules YopE, YopH, and YpkA influence endosomal MHC class II antigen processing of secreted YopE/LLO. Therefore, BMMs derived from CB6F1 mice were incubated with various Yersinia deletion mutants and used as APCs. The aim of this experiment was to compare the effect of the respective yop deletion on the relative strength of antigen presentation of the secreted versus the translocated form of LLO that served as the internal control. Infection of BMMs with wild-type Y. pseudotuberculosis strain pIB102(pHR430) translocating YopE/LLO into the host cell cytosol resulted in a pronounced stimulation of LLO190-201-specific CD4 T cells (Fig. 2A). In contrast, APCs infected with pIB102(pHR429) secreting YopE/LLO to the extracellular environment stimulated a significantly weaker antigen-specific CD4 T-cell response. Thus, results from Fig. 2A confirm previous observations (72).

FIG. 2.
  • Open in new tab
  • Download powerpoint
FIG. 2.

Influence of individual Yops on MHC class II antigen presentation of secreted and translocated hybrid YopE/LLO proteins. BMMs were infected with various Y. pseudotuberculosis mutant strains bearing deletions of individual yop genes, as indicated (see also Table 1). (A to E) MHC class II antigen presentation of secreted (pHR429) and translocated (pHR430) hybrid YopE/LLO proteins was monitored with an LLO190-201-specific I-Ab-restricted CD4 T-cell line. Cells were infected at an MOI of ∼10, ∼1, or ∼0.1, as indicated. T-cell activation was measured by determination of the amount of IFN-γ secreted into the culture supernatant. Means and standard deviations of duplicate cultures are indicated. The results of one out of four independent experiments are shown.

In contrast to the results shown in Fig. 2A, it is shown in Fig. 2B and C that at MOIs of 0.1 and 1, no significant differences in MHC class II antigen presentation were observed using Y. pseudotuberculosis ΔyopE and ΔyopH mutant strains pIB522 and pIB29 expressing secreted and translocated YopE/LLO, respectively. Interestingly, at an MOI of 10, this effect of YopE or YopH on exogenous MHC class II antigen presentation was not observed. However, by employing the ΔyopE ΔyopH double mutant strain pIB251 (Fig. 2E), the levels of CD4 T-cell activation were comparable after secretion or translocation of YopE/LLO at MOIs of 0.1, 1, and 10. As demonstrated in Fig. 2D, YpkA did not contribute to the inhibition of exogenous MHC class II antigen presentation by APCs infected with Y. pseudotuberculosis. Thus, with the exception of the ΔyopE ΔyopH double-deletion mutant, T-cell activation was always more efficient after infection with the strain translocating LLO. The difference between translocated and secreted LLO was also significantly reduced in the ΔyopE and ΔyopH single-deletion mutants, indicating that expression of these genes inhibited exogenous antigen presentation.

To verify these observations, BMMs were coincubated with purified p60 from L. monocytogenes and various Y. pseudotuberculosis strains. Similar to the results obtained after coinfection with yersiniae secreting hybrid YopE/LLO proteins, the presentation of exogenous p60 was inhibited by the Y. pseudotuberculosis wild-type strain pIB102 (Fig. 3A). This inhibitory activity was dependent on the presence of YopE and YopH, as demonstrated by the significantly improved presentation of exogenous p60 after coincubation with the ΔyopE ΔyopH double mutant strain pIB251. In contrast to native p60, the presentation of the cognate epitope p60301-312 was inhibited to a much lesser extent (Fig. 3B).

FIG. 3.
  • Open in new tab
  • Download powerpoint
FIG. 3.

Influence of individual Yops on MHC class II antigen presentation after exogenous loading of p60. BMMs were infected with various Y. pseudotuberculosis mutant strains bearing deletions of individual yop genes and were simultaneously loaded with purified p60. The presentation of the T-cell antigen p60 was monitored with an I-Ad-restricted p60301-312-specific CD4 T-cell line. T-cell activation was measured by determination of the amount of IFN-γ secreted into the culture supernatant. The absolute IFN-γ concentration (A) and the relative IFN-γ concentration (B) in relation to those for the ΔyopE ΔyopH double mutant strain pIB251 are shown. Cells loaded externally with p60301-312 were used as a control. Means and standard deviations of duplicate cultures are indicated. Asterisks indicate a significant difference compared to the control without inhibitor. The results of one out of four independent experiments are shown.

In order to visualize the influence of YopE and YopH on exogenous antigen uptake, macrophage-like P388D1 cell monolayers were incubated with fluorescently labeled OVA and with wild-type Y. pseudotuberculosis strain pIB102 or ΔyopE ΔyopH double mutant strain pIB251. Cells were fixed and processed for differential immunofluorescence staining with an antibody directed against the Yersinia LPS, as indicated in Materials and Methods. Figure 4, upper panels, shows typical images obtained by an overlay of the fluorescence signals from TRITC (extracellular Yersinia) and AMCA (intra- and extracellular Yersinia). Thus, internalized bacteria (AMCA positive and TRITC negative) appear blue, whereas extracellular yersiniae (AMCA and TRITC positive) exhibit a purple fluorescent color (mixture of blue and red). Wild-type Y. pseudotuberculosis strain pIB102 inhibited its own uptake and almost completely inhibited the engulfment of OVA-FITC by macrophage-like cells, whereas in P388D1 cells infected with pIB251, the number of endosomes labeled with OVA-FITC was comparable to that in noninfected macrophages.

FIG. 4.
  • Open in new tab
  • Download powerpoint
FIG. 4.

Effects of YopE and YopH on antigen uptake visualized by immunofluorescence microscopy. Macrophage-like P388D1 cells were incubated with fluorescently labeled OVA together with wild-type Y. pseudotuberculosis strain pIB102 or ΔyopE ΔyopH double mutant strain pIB251. Intra- and extracellular yersiniae were visualized by differential immunofluorescence. The upper panels reveal the distribution of intracellular (blue) and extracellular (purple) yersiniae, the middle panels demonstrate the distribution of fluorescently labeled ovalbumin (green) after phagocytosis, and the lower panels show macrophages (phase contrast of corresponding images). The results of one out of two independent experiments are shown.

In summary, these results indicate that the concerted action of YopE and YopH has a significant inhibitory impact on exogenous antigen uptake and MHC class II presentation.

Alternative MHC class II antigen presentation of translocated hybrid proteins is independent from YopE as a type III carrier molecule and is not restricted to LLO.In further experiments, two important questions were addressed. Does the alternative MHC class II antigen presentation of translocated hybrid LLO depend on YopE as a carrier molecule, and is this type of antigen presentation restricted to LLO? As an alternative to YopE used for cytosolic foreign antigen delivery by Yersinia, the translocated type III effector protein YopH, a tyrosine phosphatase (42), was employed. In addition to LLO, p60 was engaged as a second antigen. Thus, four plasmid vectors were constructed (Table 1). Plasmid pHR514 encodes the N-terminal 32 aa of YopH fused to residues 51 to 363 of LLO, resulting in a hybrid protein that contains the secretion domain but lacks the translocation domain of YopH. Plasmid pHR507 bears the genetic information for YopH1-138/LLO51-363. This hybrid protein contains both the secretion and translocation domains of YopH. In contrast, plasmids pHR116 and pHR119 encode the translocated and secreted YopH/p60 fusion proteins, respectively (Table 1).

Antigen presentation assays using Y. pseudotuberculosis wild-type strain pIB102 expressing the newly constructed chimeric YopH proteins yielded results comparable to those obtained from assays employing hybrid YopE proteins. MHC class II antigen presentation of LLO (Fig. 5A) as well as p60 (Fig. 5B) was dependent on YopH-mediated translocation of the respective fusion protein into the cytosol of the APCs. In contrast, pIB102 carrying plasmid pHR514 or pHR119, coding for secreted hybrid YopH1-32/LLO51-363 and YopH1-32/p60130-477 proteins, respectively, was not recognized by LLO190-201-or p60301-312-specific CD4 T cells.

FIG. 5.
  • Open in new tab
  • Download powerpoint
FIG. 5.

MHC class II antigen presentation of translocated hybrid YopH proteins. BMMs were infected with various Y. pseudotuberculosis strains expressing either a YopH/LLO or a YopH/p60 fusion protein. (A) MHC class II presentation of the secreted (pHR514) and translocated (pHR507) YopH/LLO fusion proteins was monitored with an LLO190-201-specific I-Ab-restricted CD4 T-cell line. (B) MHC class II presentation of the secreted (pHR119) and translocated (pHR116) YopH/p60 fusion proteins was monitored with a p60301-312-specific I-Ad-restricted CD4 T-cell line. Nontransformed pIB102 strains were used as controls in both experiments. Cells were infected at an MOI of ∼10, ∼1, or ∼0.1, as indicated. T-cell activation was measured by determination of the amount of IFN-γ secreted into the culture supernatant. Means and standard deviations of triplicate cultures are indicated. The results of one out of two independent experiments are shown.

Taken together these data indicate that alternative MHC class II antigen presentation of proteins translocated by the Yersinia T3SS is not restricted to a specific heterologous antigen or a specific Yop carrier molecule.

Requirement of newly synthesized MHC class II molecules for presentation of LLO190-201 derived from cytosolic YopE/LLO.Brefeldin A has been widely used to differentiate two distinct pathways of MHC class II molecule access to antigenic peptides (47, 57, 64, 81). This inhibitor of anterograde movement from the endoplasmic reticulum to the Golgi apparatus blocks presentation of epitopes by newly synthesized class II proteins in the late endosome without influencing the process of peptide editing in which peptides are displayed by recycling class II molecules in early endosomal vesicles. Brefeldin A did not significantly alter the presentation of the synthetic LLO190-201 peptide in macrophages, as was expected on the basis of the binding of the peptide to preexisting cell surface class II complexes (Fig. 6). However, brefeldin A blocked LLO190-201 presentation in macrophages infected with pIB102(pHR430), indicating that processing of translocated cytosolic YopE/LLO was dependent on the binding of the LLO190-201 epitope to newly synthesized MHC class II α/β heterodimers en route to the cell surface.

FIG. 6.
  • Open in new tab
  • Download powerpoint
FIG. 6.

Requirement of de novo-synthesized MHC class II molecules for the presentation of cytosolic hybrid YopE/LLO proteins. MHC class II antigen presentation by BMMs infected with pIB102(pHR430) at an MOI of 10 was tested in the presence of 0.1 μM brefeldin A or in the absence of the inhibitor. Control APCs were loaded with synthetic LLO190-201 peptide. At a concentration of 0.1 μM, brefeldin A is known to selectively block the anterograde movement of MHC heavy-chain molecules from the endoplasmic reticulum to the Golgi apparatus. T-cell activation was measured by determination of the amount of IFN-γ secreted into the culture supernatant. Means and standard deviations of triplicate determinations are shown. An asterisk indicates a significant difference compared to the control without inhibitor. The results of one out of two independent experiments are shown.

MHC class II presentation of translocated hybrid YopE proteins occurs via an alternative endogenous MHC class II antigen presentation pathway.We used various inhibitors of antigen processing to characterize the alternative endogenous antigen presentation pathway which allows MHC class II presentation of translocated hybrid YopE proteins in the presence of YopE/YopH-mediated inhibition of exogenous antigen presentation. For inhibition experiments, we employed the previously described Y. pseudotuberculosis yopK null mutant strain pIB155 (48) that was shown to hypertranslocate chimeric YopE/LLO (72). Enhanced presentation of hypertranslocated antigen allowed a more sensitive assessment of the activity of protease inhibitors. Processing and presentation of CD4 T-cell epitope p60177-188 or LLO190-201 after infection of macrophages with pIB155(pHR414) or pIB155(pHR430) hypertranslocating the hybrid YopE1-138/p60 and YopE1-138/LLO proteins, respectively, were very sensitive to the inhibition of endosomal acidification by chloroquin but independent of the inhibition from endosomal proteases by leupeptin (Fig. 7A and B). MHC class II antigen presentation was also not inhibited by the highly specific inhibitors of the proteasome lactacystin or epoxomycin, which selectively inhibited the presentation of the CD8 T-cell epitopes p60217-225 or LLO91-99 (Fig. 7A and B). Remarkably, MHC class II but not MHC class I antigen presentation was also significantly (∼50 to 70%) inhibited in the presence of AAF-CMK, an inhibitor of TPPII. TPPII is a large cytosolic enzyme with tripeptidyl aminopeptidase and endopeptidase activity that has been implied in proteasome-independent antigen presentation (66, 89).

FIG. 7.
  • Open in new tab
  • Download powerpoint
FIG. 7.

MHC class II presentation of cytosolic hybrid YopE/LLO proteins occurs independently of lysosomal proteases, the proteasome, and TAP. The influence of inhibitors of cytosolic (epoxomycin, lactacystin, AAF-CMK) and lysosomal (leupeptin, chloroquin) proteases on antigen presentation of cytosolic hybrid YopE/p60 (A) and YopE/LLO (B) proteins was tested in an antigen presentation assay with p60- and LLO-specific CD4 (upper panels) and CD8 (lower panels) T-cell lines. BMMs were infected with pIB155(pHR414) or pIB155(pHR430), Y. pseudotuberculosis strains hypertranslocating YopE/p60 and YopE/LLO, respectively. Antigen presentation was detected with CD4 T-cell lines directed against p60177-188 or LLO190-201 and CD8 T-cell lines directed against p60217-225 or LLO91-99, as indicated. (C) MHC class II antigen presentation by BMMs from Tap+/+ and Tap−/− mice was tested with an LLO190-201-specific CD4 T-cell line after infection with pIB102(pHR430). (D) MHC class II antigen presentation by BMMs was tested with an LLO190-201-specific CD4 T-cell line after infection with ΔyopE ΔyopH double mutant strain pIB251(pHR430) or pIB251(pHR429), which secrete and translocate YopE/LLO, respectively. Synthetic LLO190-201 peptide and L. monocytogenes SN (LM SN) were included as controls. APCs were infected at an MOI of ∼10. T-cell activation was measured by determination of the amount of IFN-γ secreted into the culture supernatant. Data are presented as the absolute IFN-γ concentration or as the percent T-cell activation compared to that for the controls in the absence of inhibitor, as indicated. Means and standard deviations of triplicate determinations are shown. Asterisks indicate a significant difference compared to the control without inhibitor. All experiments were repeated at least twice, with similar results being obtained in each one.

Efficient MHC class II presentation also occurred in macrophages derived from TAP−/− mice, indicating that endogenous antigen processing of translocated hybrid YopE proteins was TAP independent (Fig. 7C). It is important to mention that similar results were obtained when the Y. pseudotuberculosis wild-type strain pIB102 was used for inhibition experiments (data not shown).

As shown in Fig. 4, the ΔyopE ΔyopH double mutant strain pIB251 is internalized by macrophages. Therefore, it is expected that YopE1-18/LLO is secreted into the phagolysosomal vacuole and YopE1-138/LLO is translocated into the cytosol of the host cell. Similar to antigen translocated by wild-type strain pIB102 or hypertranslocating strain pIB155, MHC class II presentation of translocated YopE1-138/LLO expressed by pIB251 was not inhibited by the lysosomal protease inhibitor leupeptin (Fig. 7D), indicating that the translocated antigen is the major source of MHC class II-restricted antigen presentation after infection with a strain that is capable of antigen translocation. In contrast, MHC class II presentation of YopE1-18/LLO secreted by pIB251 was inhibited by leupeptin, indicating that antigen secreted by Y. pseudotuberculosis principally can be presented via the endosomal MHC class II antigen presentation pathway. This result clearly indicates that translocated hybrid YopE proteins enter a distinctive alternative MHC class II antigen presentation pathway that is not affected by YopE/YopH-mediated inhibition of exogenous antigen uptake.

Requirement of autophagy for presentation of LLO190-201 derived from cytosolic YopE/LLO.Endogenous MHC class II processing of cytosolic antigen is a long-known phenomenon and has been demonstrated for cytosolic cellular proteins as well as for various viral antigens (29, 51, 65, 70, 71). More recently, intracellular autophagy was defined as a pathway that delivers cytosolic antigens for further presentation in the context of MHC class II molecules (27, 30, 56, 60, 91). Two different pathways have been defined. While macroautophagy requires processing in the phagolysosomal compartment (56, 60), in chaperone-mediated autophagy, antigens are processed in the cytoplasm (91). The macroautophagy pathway has been characterized in detail in cells transfected with the cytosolic model antigen neomycin phosphotransferase II (NeoR) (56) as well as in cells latently infected with Epstein-Barr virus (EBV) (60). In contrast to chaperone-mediated autophagy, macroautophagy can be inhibited by the inhibitors wortmannin and 3-MA. Thus, the experimental characteristic of the macroautophagy pathway is that it is equally sensitive to inhibitors of lysosomal antigen presentation and the macroautophagy inhibitors (56, 60). Therefore, in order to investigate the possible involvement of macroautophagy in the presentation of translocated hybrid YopE/LLO, we tested the effects of two inhibitors of macroautophagy, 3-MA and wortmannin, on MHC class II antigen presentation. Each of the inhibitors was added 1 h before infection of BMMs with pIB102(pHR430) or before external loading with concentrated L. monocytogenes culture supernatant as a control antigen. As shown in Fig. 8, both 3-MA and wortmannin significantly suppressed the recognition of the translocated hybrid YopE1-138/LLO protein but did not affect the presentation of externally loaded antigen.

FIG. 8.
  • Open in new tab
  • Download powerpoint
FIG. 8.

Inhibitors of the cellular autophagy pathway inhibit MHC class II presentation of translocated bacterial antigens. MHC class II antigen presentation by BMMs infected with pIB102(pHR430) was tested in the presence of 3-MA or wortmannin or in the absence of inhibitor. As a control antigen, L. monocytogenes SN (LM SN) was used for the external loading of BMMs. T-cell activation of an LLO190-201-specific CD4 T-cell line was measured by determination of the amount of IFN-γ secreted into the culture supernatant. Means and standard deviations of triplicate determinations are shown. Asterisks indicate a significant difference compared to the control without inhibitor. The results of one out of two independent experiments are shown.

To rule out any interference of the chemical inhibitors with the amount of proteins translocated into the cytosol of macrophages, BMMs were infected with Yersinia pIB102(pHR430) in the presence of all individual inhibitors used in this study. As shown in Fig. 9A and B, the inhibitors did not influence either the amount of YopE/LLO production by the bacteria (whole-cell lysates of non-cell-associated bacteria) or the amount of antigen translocated into BMMs (Triton X-100-soluble BMM lysates). It is conceivable that the treatment with 0.1% Triton X-100 results in a permeabilization of the bacteria, and therefore, the Triton X-100-soluble BMM lysate could include YopE/LLO in the bacteria internalized by these cells. Thus, YopE/LLO may not have been translocated into BMMs, as shown in Fig. 9B. To rule out this effect, a Y. pseudotuberculosis yopB mutant strain (pIB604) was used as a control. This strain is deficient in translocation of Yops but still synthesizes these effector proteins (44). BMMs were infected with Yersinia wild-type pIB102(pHR430) or pIB604(pHR430). As shown in Fig. 9C, both strains produced comparable amounts of chimeric YopE/LLO (whole-cell lysates of non-cell-associated bacteria). However, in contrast to BMMs infected with wild-type Yersinia, YopE/LLO was not detected in the Triton X-100-soluble BMM lysate from cells incubated with the yopB mutant strain (Fig. 9D).

FIG. 9.
  • Open in new tab
  • Download powerpoint
FIG. 9.

(A and B) Influence of chemical inhibitors of antigen presentation on expression and translocation of hybrid YopE/LLO. BMMs were incubated with pIB102(pHR430). Whole-cell lysates of non-cell-associated bacteria (A) and Triton X-100-soluble BMM lysates containing translocated proteins (B) were prepared and examined for the presence of chimeric YopE/LLO by Western blot analysis, as described in Materials and Methods. This experiment was repeated twice without detection of any significant effects of the chemical inhibitors on the expression and translocation of the hybrid YopE/LLO. (C and D) Specificity of the translocation assay. To rule out interference of Triton X-100 with the detection of YopE/LLO in the Triton X-100-soluble BMM lysate, a translocation-deficient Y. pseudotuberculosis yopB mutant strain (pIB604) was used a control. BMMs were incubated with pIB102(pHR430) or pIB604(pHR430). Whole-cell lysates of non-cell-associated bacteria (C) and Triton X-100-soluble BMM lysates containing translocated proteins (D) were prepared and examined for the presence of chimeric YopE/LLO by Western blot analysis, as described in Materials and Methods. This experiment was repeated twice, with identical results being obtained in each one.

In summary, the inhibition of the MHC class II-restricted presentation of translocated antigen by two inhibitors of macroautophagy and by the inhibition of lysosomal acidification together indicate that MHC class II presentation of heterologous proteins translocated by the Y. pseudotuberculosis T3SS into the cytosol of APCs depends on an alternative macroautophagy-dependent antigen presentation pathway.

DISCUSSION

In the mouse infection model, enteropathogenic yersiniae invade Peyer's patches by entering through specialized epithelial cells called M cells (41, 46). Subsequently, yersiniae disseminate to the lymph nodes, spleen, and liver, where they form microcolonies/microabscesses (5, 17). In contrast to other enteric pathogens such as Salmonella and Shigella, Yersinia is predominantly an extracellular pathogen (46, 77). Initially innate host defenses, such as polymorphonuclear leukocytes, macrophages, and natural killer cells, are involved in controlling Yersinia infection (3, 18, 50), but subsequently, a robust adaptive immune response is required to overcome Yersinia infections. Specific antibodies (86) as well as IFN-γ-producing CD4 and CD8 T cells (2, 4, 5, 13, 32) play an essential role in clearing Yersinia infections and have been shown to mediate protection in adoptive transfer experiments (2, 4, 5). It has been demonstrated that infection of rats with Y. pseudotuberculosis leads to the induction of an MHC class I-restricted cytotoxic CD8 T-cell response, but the antigens that are presented to CD8 T cells in an MHC class I context are unknown (32). However, cells infected with Yersinia have been shown to present an epitope of YopH to MHC class I-restricted CD8 T cells (80).

In the current report, we demonstrate for the first time that the concerted action of YopE and YopH from Y. pseudotuberculosis blocks the uptake and MHC class II presentation of secreted bacterial antigens. Remarkably, macrophages infected with Y. pseudotuberculosis still present epitopes in the context of MHC class II molecules after translocation of antigenic proteins into the cytoplasm of APCs by the Yersinia T3SS. This MHC class II antigen presentation of translocated proteins clearly occurs via an alternative processing pathway that involves macroautophagy and requires lysosmal acidification but is independent of the proteasome and TAP.

A number of previous studies have addressed the effect of Yops on the uptake (12, 33, 40, 68) and presentation (6) of exogenously loaded model antigens. In the present study, we extend these previous observations by directly demonstrating the effects of YopE and YopH from Y. pseudotuberculosis on the presentation of bacterially secreted model antigens. The synergistic suppressive effect of YopE and YopH on antigen uptake and presentation also confirms the findings of a previous study that suggest that Y. enterocolitica inhibits antigen degradation by dendritic cells through the interaction of multiple Yops (1). While YopE inhibits phagocytosis by its Rho GTPase-inactivating activity (10, 69, 87), the protein tyrosine phosphatase activity of YopH counteracts phagocytosis by the inhibition of early steps in the β1 integrin signaling pathway (85). It can be assumed that simultaneous inhibition of multiple RhoA activation pathways is required in order to obtain a significant inhibition of phagocytosis. However, the transport of de novo-synthesized MHC class II molecules from the endoplasmic reticulum to the phagosomal compartment and further peptide loading were not affected by YopE and YopH.

To date, endogenous MHC class II processing has been reported for cytosolic cellular proteins as well as for various viral antigens (29, 51, 65, 70, 71). A novel finding of our current study is that this immune mechanism also applies to bacterial antigens translocated into the cytoplasm of infected macrophages. MHC class II antigen presentation of cytosolic bacterial proteins is reported for two independent listerial T-cell antigens (LLO and p60) and for two different Yersinia effector proteins (YopE and YopH) used as translocated carrier proteins. Furthermore, we show that the antigen presentation pathway involved in MHC class II presentation of translocated hybrid proteins was independent of the proteasome and TAP but was inhibited by alkalinization of the phagolysosome or by blockade of the macroautophagy pathway through the action of 3-MA and wortmannin. It is known that autophagy plays an important role for the innate immune defense against intracellular bacteria such as Mycobacteria, Salmonella, and Shigella (8, 26, 43, 55, 59). In a more recent study, Deuretzbacher and colleagues analyzed the effects of Y. enterocolitica on autophagy in macrophages (28). The authors showed that autophagy was mediated by the Yersinia adhesins invasin and YadA and particularly depended on the engagement of β1 integrin receptors. Several autophagy-related events followed β1 integrin-mediated engulfment of the bacteria, including the formation of autophagosomes, processing of the marker protein LC3, redistribution of green fluorescent protein-LC3 to bacteria-containing vacuoles, and the segregation of intracellular bacteria by autophagosomal compartments. However, owing to the importance of autophagy as a host defense response, wild-type Y. enterocolitica suppressed autophagy by mobilizing type III secretion (28). It was speculated that the subversion of autophagy may be part of the Y. enterocolitica virulence strategy that supports bacterial survival when β1 integrin-dependent internalization and autophagy activation by macrophages are deleterious for the pathogen (28). Our data now provide a direct link between macroautophagy and acquired immunity against obligate extracellular yersiniae which translocate Yops into the cytoplasm of infected cells by means of their T3SSs. Obviously, suppression of autophagy during the initial interaction of macrophages and Yersinia by the concerted activities of Yops (28) does not lead to a complete impairment of autophagy. Strikingly, translocated Yops promote subversion of phagocytosis and autophagy on the one hand but are also protein substrates of remaining levels of autophagy on the other hand.

The link between macroautophagy and endogenous MHC class II antigen presentation was demonstrated in a number of recent publications (27, 30, 56, 60), and it is now also known that macroautophagy represents a process that is constitutively active in various MHC class II-expressing cells, including dendritic cells and macrophages (75). Nimmerjahn et al. have shown that endogenous presentation of an epitope derived from the cytosolic model antigen NeoR on MHC class II is mediated by autophagy and requires lysosomal processing (56). Similarly, studies with cells latently infected with EBV have shown the requirement of an autophagy- and lysosome-dependent pathway for the MHC class II-restricted presentation of EBV nuclear antigen (60). Our data clearly characterize the alternative MHC class II antigen presentation pathway of translocated chimeric YopE as being dependent on macroautophagy and lysosomal acidification. However, as MHC class II-restricted presentation of translocated hybrid YopE was not inhibited by the inhibitor of lysosomal proteases, leupeptin, we cannot exclude the possibility that antigen processing occurred independently of lysosomal processing and that phagolysosomal acidification was required just for peptide loading on MHC class II molecules in the phagolysosome. Interestingly, we present evidence that TPPII (38), a cytoplasmic protease, might be involved in endogenous MHC class II presentation of translocated chimeric YopE fusion proteins. The involvement of TPPII in cytoplasmic MHC class I processing and presentation has been the matter of some debate (7, 34, 66, 76, 89). Despite the suppressive effect of TPPII inhibition on endogenous MHC class II presentation of cytosolic hybrid YopE, we were not able to demonstrate any influence of inhibitors of the proteasome on this processing pathway. In control experiments, however, proteasome inhibitors clearly inhibited the MHC class I-restricted presentation of epitopes derived from the same YopE fusion protein. The requirement of cytoplasmic antigen processing for endogenous MHC class II antigen presentation of chimeric YopE is a characteristic that also plays a central role in the chaperone-mediated autophagy-dependent MHC class II presentation pathway described by Zhou et al. (91) and in the endogenous MHC class II presentation pathway described by Lich et al. (51), both of which involve cytoplasmic antigen presentation by the proteasome. Interestingly, compared to macroautophagy, the chaperone-dependent pathway is active in APCs showing diminished levels of macroautophagy (25, 54, 82, 90).

In the current study, we have used LLO and p60 as bacterial model antigens fused to N-terminal secretion or translocation domains of YopE or YopH in order to study the processing and presentation of defined CD4 T-cell epitopes. The inhibition of exogenous antigen presentation of these hybrid proteins after infection of cells with Y. pseudotuberculosis and the presence of an alternative endogenous MHC class II antigen presentation pathway lead to the prediction that translocated wild-type Yops bear possible CD4 T-cell epitopes and are excellent CD4 T-cell antigens. The employment of the autophagy pathway enables the host to mount an MHC class II-restricted CD4 T-cell response even in the presence of microbial immunoevasive mechanisms which suppress the presentation of microbial antigens by the exogenous antigen presentation pathway. In addition, the presence of an alternative MHC class II antigen presentation pathway for translocated bacterial Yops explains why vaccination using the Yersinia T3SS as an antigen delivery system is an excellent tool for CD8 and CD4 T-cell priming (72, 74).

ACKNOWLEDGMENTS

We thank S. Schenk for expert technical assistance.

This study was supported by Deutsche Forschungsgemeinschaft (DFG) grants RU 838/2-2 (to H. Rüssmann, B. Köhn, and S. Jellbauer) and GE 1081/2-2 (G. Geginat).

FOOTNOTES

    • Received 13 February 2010.
    • Returned for modification 17 March 2010.
    • Accepted 14 September 2010.
  • Copyright © 2010 American Society for Microbiology

REFERENCES

  1. 1.↵
    Adkins, I., M. Koberle, S. Grobner, S. E. Autenrieth, E. Bohn, S. Borgmann, and I. B. Autenrieth. 2008. Y. enterocolitica inhibits antigen degradation in dendritic cells. Microbes Infect.10:798-806.
    OpenUrlCrossRefPubMed
  2. 2.↵
    Autenrieth, I. B., M. Beer, E. Bohn, S. H. Kaufmann, and J. Heesemann. 1994. Immune responses to Yersinia enterocolitica in susceptible BALB/c and resistant C57BL/6 mice: an essential role for gamma interferon. Infect. Immun.62:2590-2599.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    Autenrieth, I. B., V. Kempf, T. Sprinz, S. Preger, and A. Schnell. 1996. Defense mechanisms in Peyer's patches and mesenteric lymph nodes against Yersinia enterocolitica involve integrins and cytokines. Infect. Immun.64:1357-1368.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    Autenrieth, I. B., A. Tingle, A. Reske-Kunz, and J. Heesemann. 1992. T lymphocytes mediate protection against Yersinia enterocolitica in mice: characterization of murine T-cell clones specific for Y. enterocolitica. Infect. Immun.60:1140-1149.
    OpenUrlAbstract/FREE Full Text
  5. 5.↵
    Autenrieth, I. B., U. Vogel, S. Preger, B. Heymer, and J. Heesemann. 1993. Experimental Yersinia enterocolitica infection in euthymic and T-cell-deficient athymic nude C57BL/6 mice: comparison of time course, histomorphology, and immune response. Infect. Immun.61:2585-2595.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    Autenrieth, S. E., I. Soldanova, R. Rosemann, D. Gunst, N. Zahir, M. Kracht, K. Ruckdeschel, H. Wagner, S. Borgmann, and I. B. Autenrieth. 2007. Yersinia enterocolitica YopP inhibits MAP kinase-mediated antigen uptake in dendritic cells. Cell. Microbiol.9:425-437.
    OpenUrlCrossRefPubMed
  7. 7.↵
    Basler, M., and M. Groettrup. 2007. No essential role for tripeptidyl peptidase II for the processing of LCMV-derived T cell epitopes. Eur. J. Immunol.37:896-904.
    OpenUrlCrossRefPubMed
  8. 8.↵
    Birmingham, C. L., A. C. Smith, M. A. Bakowski, T. Yoshimori, and J. H. Brumell. 2006. Autophagy controls Salmonella infection in response to damage to the Salmonella-containing vacuole. J. Biol. Chem.281:11374-11383.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    Black, D. S., and J. B. Bliska. 1997. Identification of p130Cas as a substrate of Yersinia YopH (Yop51), a bacterial protein tyrosine phosphatase that translocates into mammalian cells and targets focal adhesions. EMBO J.16:2730-2744.
    OpenUrlAbstract
  10. 10.↵
    Black, D. S., and J. B. Bliska. 2000. The RhoGAP activity of the Yersinia pseudotuberculosis cytotoxin YopE is required for antiphagocytic function and virulence. Mol. Microbiol.37:515-527.
    OpenUrlCrossRefPubMedWeb of Science
  11. 11.↵
    Black, D. S., A. Marie-Cardine, B. Schraven, and J. B. Bliska. 2000. The Yersinia tyrosine phosphatase YopH targets a novel adhesion-regulated signalling complex in macrophages. Cell. Microbiol.2:401-414.
    OpenUrlCrossRefPubMedWeb of Science
  12. 12.↵
    Bliska, J. B., and D. S. Black. 1995. Inhibition of the Fc receptor-mediated oxidative burst in macrophages by the Yersinia pseudotuberculosis tyrosine phosphatase. Infect. Immun.63:681-685.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    Bohn, E., and I. B. Autenrieth. 1996. IL-12 is essential for resistance against Yersinia enterocolitica by triggering IFN-gamma production in NK cells and CD4+ T cells. J. Immunol.156:1458-1468.
    OpenUrlAbstract
  14. 14.
    Bölin, I., and H. Wolf-Watz. 1984. Molecular cloning of the temperature-inducible outer membrane protein 1 of Yersinia pseudotuberculosis. Infect. Immun.43:72-78.
    OpenUrlAbstract/FREE Full Text
  15. 15.
    Bölin, I., and H. Wolf-Watz. 1988. The virulence plasmid-encoded Yop2b protein of Yersinia pseudotuberculosis is a virulence determinant regulated by calcium and temperature at transcriptional level. Mol. Microbiol.2:237-245.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    Braciale, T. J., L. A. Morrison, M. T. Sweetser, J. Sambrook, M. J. Gething, and V. L. Braciale. 1987. Antigen presentation pathways to class I and class II MHC-restricted T lymphocytes. Immunol. Rev.98:95-114.
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.↵
    Carter, P. B. 1975. Pathogenicity of Yersinia enterocolitica for mice. Infect. Immun.11:164-170.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    Carter, P. B., T. T. MacDonald, and F. M. Collins. 1979. Host responses to infection with Yersinia enterocolitica. Contrib. Microbiol. Immunol.5:346-350.
    OpenUrlPubMed
  19. 19.↵
    Chiang, H. L., S. R. Terlecky, C. P. Plant, and J. F. Dice. 1989. A role for a 70-kilodalton heat shock protein in lysosomal degradation of intracellular proteins. Science246:382-385.
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    Chicz, R. M., R. G. Urban, J. C. Gorga, D. A. Vignali, W. S. Lane, and J. L. Strominger. 1993. Specificity and promiscuity among naturally processed peptides bound to HLA-DR alleles. J. Exp. Med.178:27-47.
    OpenUrlAbstract/FREE Full Text
  21. 21.↵
    Cornelis, G. R. 2006. The type III secretion injectisome. Nat. Rev. Microbiol.4:811-825.
    OpenUrlCrossRefPubMedWeb of Science
  22. 22.↵
    Cornelis, G. R. 2002. The Yersinia Ysc-Yop ‘type III’ weaponry. Nat. Rev. Mol. Cell Biol.3:742-752.
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.↵
    Cuervo, A. M., and J. F. Dice. 1996. A receptor for the selective uptake and degradation of proteins by lysosomes. Science273:501-503.
    OpenUrlAbstract
  24. 24.↵
    Cuervo, A. M., and J. F. Dice. 2000. Unique properties of lamp2a compared to other lamp2 isoforms. J. Cell Sci.113(Pt. 24):4441-4450.
    OpenUrlAbstract/FREE Full Text
  25. 25.↵
    Dani, A., A. Chaudhry, P. Mukherjee, D. Rajagopal, S. Bhatia, A. George, V. Bal, S. Rath, and S. Mayor. 2004. The pathway for MHC II-mediated presentation of endogenous proteins involves peptide transport to the endo-lysosomal compartment. J. Cell Sci.117:4219-4230.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    de Chastellier, C., and L. Thilo. 2006. Cholesterol depletion in Mycobacterium avium-infected macrophages overcomes the block in phagosome maturation and leads to the reversible sequestration of viable mycobacteria in phagolysosome-derived autophagic vacuoles. Cell. Microbiol.8:242-256.
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.↵
    Dengjel, J., O. Schoor, R. Fischer, M. Reich, M. Kraus, M. Muller, K. Kreymborg, F. Altenberend, J. Brandenburg, H. Kalbacher, R. Brock, C. Driessen, H. G. Rammensee, and S. Stevanovic. 2005. Autophagy promotes MHC class II presentation of peptides from intracellular source proteins. Proc. Natl. Acad. Sci. U. S. A.102:7922-7927.
    OpenUrlAbstract/FREE Full Text
  28. 28.↵
    Deuretzbacher, A., N. Czymmeck, R. Reimer, K. Trülzsch, K. Gaus, H. Hohenberg, J. Heesemann, M. Aepfelbacher, and K. Ruckdeschel. 2009. Beta1 integrin-dependent engulfment of Yersinia enterocolitica by macrophages is coupled to the activation of autophagy and suppressed by type III protein secretion. J. Immunol.183:5847-5860.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    Dongre, A. R., S. Kovats, P. deRoos, A. L. McCormack, T. Nakagawa, V. Paharkova-Vatchkova, J. Eng, H. Caldwell, J. R. Yates III, and A. Y. Rudensky. 2001. In vivo MHC class II presentation of cytosolic proteins revealed by rapid automated tandem mass spectrometry and functional analyses. Eur. J. Immunol.31:1485-1494.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    Dorfel, D., S. Appel, F. Grunebach, M. M. Weck, M. R. Muller, A. Heine, and P. Brossart. 2005. Processing and presentation of HLA class I and II epitopes by dendritic cells after transfection with in vitro-transcribed MUC1 RNA. Blood105:3199-3205.
    OpenUrlAbstract/FREE Full Text
  31. 31.↵
    Dukuzumuremyi, J. M., R. Rosqvist, B. Hallberg, B. Akerstrom, H. Wolf-Watz, and K. Schesser. 2000. The Yersinia protein kinase A is a host factor inducible RhoA/Rac-binding virulence factor. J. Biol. Chem.275:35281-35290.
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    Falgarone, G., H. S. Blanchard, F. Virecoulon, M. Simonet, and M. Breban. 1999. Coordinate involvement of invasin and Yop proteins in a Yersinia pseudotuberculosis-specific class I-restricted cytotoxic T cell-mediated response. J. Immunol.162:2875-2883.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    Fallman, M., K. Andersson, S. Hakansson, K. E. Magnusson, O. Stendahl, and H. Wolf-Watz. 1995. Yersinia pseudotuberculosis inhibits Fc receptor-mediated phagocytosis in J774 cells. Infect. Immun.63:3117-3124.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    Firat, E., J. Huai, L. Saveanu, S. Gaedicke, P. Aichele, K. Eichmann, P. van Endert, and G. Niedermann. 2007. Analysis of direct and cross-presentation of antigens in TPPII knockout mice. J. Immunol.179:8137-8145.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    Galan, J. E., and H. Wolf-Watz. 2006. Protein delivery into eukaryotic cells by type III secretion machines. Nature444:567-573.
    OpenUrlCrossRefPubMedWeb of Science
  36. 36.↵
    Geginat, G., M. Lalic, M. Kretschmar, W. Goebel, H. Hof, D. Palm, and A. Bubert. 1998. Th1 cells specific for a secreted protein of Listeria monocytogenes are protective in vivo. J. Immunol.160:6046-6055.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    Geginat, G., S. Schenk, M. Skoberne, W. Goebel, and H. Hof. 2001. A novel approach of direct ex vivo epitope mapping identifies dominant and subdominant CD4 and CD8 T cell epitopes from Listeria monocytogenes. J. Immunol.166:1877-1884.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    Geier, E., G. Pfeifer, M. Wilm, M. Lucchiari-Hartz, W. Baumeister, K. Eichmann, and G. Niedermann. 1999. A giant protease with potential to substitute for some functions of the proteasome. Science283:978-981.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    Germain, R. N., and D. H. Margulies. 1993. The biochemistry and cell biology of antigen processing and presentation. Annu. Rev. Immunol.11:403-450.
    OpenUrlCrossRefPubMedWeb of Science
  40. 40.↵
    Grosdent, N., I. Maridonneau-Parini, M. P. Sory, and G. R. Cornelis. 2002. Role of Yops and adhesins in resistance of Yersinia enterocolitica to phagocytosis. Infect. Immun.70:4165-4176.
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    Grutzkau, A., C. Hanski, H. Hahn, and E. O. Riecken. 1990. Involvement of M cells in the bacterial invasion of Peyer's patches: a common mechanism shared by Yersinia enterocolitica and other enteroinvasive bacteria. Gut31:1011-1015.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    Guan, K. L., and J. E. Dixon. 1990. Protein tyrosine phosphatase activity of an essential virulence determinant in Yersinia. Science249:553-556.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    Gutierrez, M. G., S. S. Master, S. B. Singh, G. A. Taylor, M. I. Colombo, and V. Deretic. 2004. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell119:753-766.
    OpenUrlCrossRefPubMedWeb of Science
  44. 44.↵
    Hakansson, S., K. Schesser, C. Persson, E. E. Galyov, R. Rosqvist, F. Homble, and H. Wolf-Watz. 1996. The YopB protein of Yersinia pseudotuberculosis is essential for the translocation of Yop effector proteins across the target cell plasma membrane and displays a contact-dependent membrane disrupting activity. EMBO J.15:5812-5823.
    OpenUrlPubMedWeb of Science
  45. 45.↵
    Hamid, N., A. Gustavsson, K. Andersson, K. McGee, C. Persson, C. E. Rudd, and M. Fallman. 1999. YopH dephosphorylates Cas and Fyn-binding protein in macrophages. Microb. Pathog.27:231-242.
    OpenUrlCrossRefPubMedWeb of Science
  46. 46.↵
    Hanski, C., U. Kutschka, H. P. Schmoranzer, M. Naumann, A. Stallmach, H. Hahn, H. Menge, and E. O. Riecken. 1989. Immunohistochemical and electron microscopic study of interaction of Yersinia enterocolitica serotype O8 with intestinal mucosa during experimental enteritis. Infect. Immun.57:673-678.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    Harding, C. V. 1996. Class II antigen processing: analysis of compartments and functions. Crit. Rev. Immunol.16:13-29.
    OpenUrlCrossRefPubMedWeb of Science
  48. 48.↵
    Holmström, A., J. Petterson, R. Rosqvist, S. Hakansson, F. Tafazoli, M. Fallman, K. E. Magnusson, H. Wolf-Watz, and A. Forsberg. 1997. YopK of Yersinia pseudotuberculosis controls translocation of Yop effectors across the eukaryotic cell membrane. Mol. Microbiol.24:73-91.
    OpenUrlCrossRefPubMedWeb of Science
  49. 49.↵
    Juris, S. J., A. E. Rudolph, D. Huddler, K. Orth, and J. E. Dixon. 2000. A distinctive role for the Yersinia protein kinase: actin binding, kinase activation, and cytoskeleton disruption. Proc. Natl. Acad. Sci. U. S. A.97:9431-9436.
    OpenUrlAbstract/FREE Full Text
  50. 50.↵
    Kerschen, E. J., D. A. Cohen, A. M. Kaplan, and S. C. Straley. 2004. The plague virulence protein YopM targets the innate immune response by causing a global depletion of NK cells. Infect. Immun.72:4589-4602.
    OpenUrlAbstract/FREE Full Text
  51. 51.↵
    Lich, J. D., J. F. Elliott, and J. S. Blum. 2000. Cytoplasmic processing is a prerequisite for presentation of an endogenous antigen by major histocompatibility complex class II proteins. J. Exp. Med.191:1513-1524.
    OpenUrlAbstract/FREE Full Text
  52. 52.↵
    Malnati, M. S., S. Ceman, M. Weston, R. DeMars, and E. O. Long. 1993. Presentation of cytosolic antigen by HLA-DR requires a function encoded in the class II region of the MHC. J. Immunol.151:6751-6756.
    OpenUrlAbstract
  53. 53.↵
    Malnati, M. S., M. Marti, T. LaVaute, D. Jaraquemada, W. Biddison, R. DeMars, and E. O. Long. 1992. Processing pathways for presentation of cytosolic antigen to MHC class II-restricted T cells. Nature357:702-704.
    OpenUrlCrossRefPubMed
  54. 54.↵
    Massey, A. C., S. Kaushik, G. Sovak, R. Kiffin, and A. M. Cuervo. 2006. Consequences of the selective blockage of chaperone-mediated autophagy. Proc. Natl. Acad. Sci. U. S. A.103:5805-5810.
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    Nakagawa, I., A. Amano, N. Mizushima, A. Yamamoto, H. Yamaguchi, T. Kamimoto, A. Nara, J. Funao, M. Nakata, K. Tsuda, S. Hamada, and T. Yoshimori. 2004. Autophagy defends cells against invading group A Streptococcus. Science306:1037-1040.
    OpenUrlAbstract/FREE Full Text
  56. 56.↵
    Nimmerjahn, F., S. Milosevic, U. Behrends, E. M. Jaffee, D. M. Pardoll, G. W. Bornkamm, and J. Mautner. 2003. Major histocompatibility complex class II-restricted presentation of a cytosolic antigen by autophagy. Eur. J. Immunol.33:1250-1259.
    OpenUrlCrossRefPubMedWeb of Science
  57. 57.↵
    Nuchtern, J. G., W. E. Biddison, and R. D. Klausner. 1990. Class II MHC molecules can use the endogenous pathway of antigen presentation. Nature343:74-76.
    OpenUrlCrossRefPubMed
  58. 58.↵
    Obert, S., R. J. O'Connor, S. Schmid, and P. Hearing. 1994. The adenovirus E4-6/7 protein transactivates the E2 promoter by inducing dimerization of a heteromeric E2F complex. Mol. Cell. Biol.14:1333-1346.
    OpenUrlAbstract/FREE Full Text
  59. 59.↵
    Ogawa, M., T. Yoshimori, T. Suzuki, H. Sagara, N. Mizushima, and C. Sasakawa. 2005. Escape of intracellular Shigella from autophagy. Science307:727-731.
    OpenUrlAbstract/FREE Full Text
  60. 60.↵
    Paludan, C., D. Schmid, M. Landthaler, M. Vockerodt, D. Kube, T. Tuschl, and C. Munz. 2005. Endogenous MHC class II processing of a viral nuclear antigen after autophagy. Science307:593-596.
    OpenUrlAbstract/FREE Full Text
  61. 61.↵
    Paschen, A., M. Song, S. Schenk, J. Janda, X. D. Nguyen, W. Osen, D. Schadendorf, and G. Geginat. 2006. Identification of a cross-reactive HLA-DRB1*0301-restricted CD4 T cell response directed against cholesterol-binding cytolysins from two different pathogens. Microbes Infect.8:2034-2043.
    OpenUrlCrossRefPubMed
  62. 62.↵
    Persson, C., N. Carballeira, H. Wolf-Watz, and M. Fallman. 1997. The PTPase YopH inhibits uptake of Yersinia, tyrosine phosphorylation of p130Cas and FAK, and the associated accumulation of these proteins in peripheral focal adhesions. EMBO J.16:2307-2318.
    OpenUrlAbstract
  63. 63.↵
    Pieters, J. 2000. MHC class II-restricted antigen processing and presentation. Adv. Immunol.75:159-208.
    OpenUrlCrossRefPubMedWeb of Science
  64. 64.↵
    Pinet, V., M. S. Malnati, and E. O. Long. 1994. Two processing pathways for the MHC class II-restricted presentation of exogenous influenza virus antigen. J. Immunol.152:4852-4860.
    OpenUrlAbstract
  65. 65.↵
    Qi, L., J. M. Rojas, and S. Ostrand-Rosenberg. 2000. Tumor cells present MHC class II-restricted nuclear and mitochondrial antigens and are the predominant antigen presenting cells in vivo. J. Immunol.165:5451-5461.
    OpenUrlAbstract/FREE Full Text
  66. 66.↵
    Reits, E., J. Neijssen, C. Herberts, W. Benckhuijsen, L. Janssen, J. W. Drijfhout, and J. Neefjes. 2004. A major role for TPPII in trimming proteasomal degradation products for MHC class I antigen presentation. Immunity20:495-506.
    OpenUrlCrossRefPubMed
  67. 67.
    Rosqvist, R., M. Skurnik, and H. Wolf-Watz. 1988. Increased virulence of Yersinia pseudotuberculosis by two independent mutations. Nature334:522-524.
    OpenUrlCrossRefPubMed
  68. 68.↵
    Rosqvist, R., A. Forsberg, M. Rimpilainen, T. Bergman, and H. Wolf-Watz. 1990. The cytotoxic protein YopE of Yersinia obstructs the primary host defence. Mol. Microbiol.4:657-667.
    OpenUrlCrossRefPubMedWeb of Science
  69. 69.↵
    Rosqvist, R., A. Forsberg, and H. Wolf-Watz. 1991. Intracellular targeting of the Yersinia YopE cytotoxin in mammalian cells induces actin microfilament disruption. Infect. Immun.59:4562-4569.
    OpenUrlAbstract/FREE Full Text
  70. 70.↵
    Rudensky, A., P. Preston-Hurlburt, S. C. Hong, A. Barlow, and C. A. Janeway, Jr. 1991. Sequence analysis of peptides bound to MHC class II molecules. Nature353:622-627.
    OpenUrlCrossRefPubMed
  71. 71.↵
    Rudensky, A. Y., S. M. Mazel, and V. L. Yurin. 1990. Presentation of endogenous immunoglobulin determinant to immunoglobulin-recognizing T cell clones by the thymic cells. Eur. J. Immunol.20:2235-2239.
    OpenUrlCrossRefPubMed
  72. 72.↵
    Rüssmann, H., U. Gerdemann, E. I. Igwe, K. Panthel, J. Heesemann, S. Garbom, H. Wolf-Watz, and G. Geginat. 2003. Attenuated Yersinia pseudotuberculosis carrier vaccine for simultaneous antigen-specific CD4 and CD8 T-cell induction. Infect. Immun.71:3463-3472.
    OpenUrlAbstract/FREE Full Text
  73. 73.↵
    Rüssmann, H., E. I. Igwe, J. Sauer, W. D. Hardt, A. Bubert, and G. Geginat. 2001. Protection against murine listeriosis by oral vaccination with recombinant Salmonella expressing hybrid Yersinia type III proteins. J. Immunol.167:357-365.
    OpenUrlAbstract/FREE Full Text
  74. 74.↵
    Rüssmann, H., A. Weissmüller, G. Geginat, E. I. Igwe, A. Roggenkamp, A. Bubert, W. Goebel, H. Hof, and J. Heesemann. 2000. Yersinia enterocolitica-mediated translocation of defined fusion proteins to the cytosol of mammalian cells results in peptide-specific MHC class I-restricted antigen presentation. Eur. J. Immunol.30:1375-1384.
    OpenUrlCrossRefPubMed
  75. 75.↵
    Schmid, D., M. Pypaert, and C. Munz. 2007. Antigen-loading compartments for major histocompatibility complex class II molecules continuously receive input from autophagosomes. Immunity26:79-92.
    OpenUrlCrossRefPubMedWeb of Science
  76. 76.↵
    Seifert, U., C. Maranon, A. Shmueli, J. F. Desoutter, L. Wesoloski, K. Janek, P. Henklein, S. Diescher, M. Andrieu, H. de la Salle, T. Weinschenk, H. Schild, D. Laderach, A. Galy, G. Haas, P. M. Kloetzel, Y. Reiss, and A. Hosmalin. 2003. An essential role for tripeptidyl peptidase in the generation of an MHC class I epitope. Nat. Immunol.4:375-379.
    OpenUrlCrossRefPubMedWeb of Science
  77. 77.↵
    Simonet, M., S. Richard, and P. Berche. 1990. Electron microscopic evidence for in vivo extracellular localization of Yersinia pseudotuberculosis harboring the pYV plasmid. Infect. Immun.58:841-845.
    OpenUrlAbstract/FREE Full Text
  78. 78.↵
    Skoberne, M., S. Schenk, H. Hof, and G. Geginat. 2002. Cross-presentation of Listeria monocytogenes-derived CD4 T cell epitopes. J. Immunol.169:1410-1418.
    OpenUrlAbstract/FREE Full Text
  79. 79.↵
    Sory, M. P., A. Boland, I. Lambermont, and G. R. Cornelis. 1995. Identification of the YopE and YopH domains required for secretion and internalization into the cytosol of macrophages, using the cyaA gene fusion approach. Proc. Natl. Acad. Sci. U. S. A.92:1998-2002.
    OpenUrlAbstract/FREE Full Text
  80. 80.↵
    Starnbach, M. N., and M. J. Bevan. 1994. Cells infected with Yersinia present an epitope to class I MHC-restricted CTL. J. Immunol.153:1603-1612.
    OpenUrlAbstract/FREE Full Text
  81. 81.↵
    St-Pierre, Y., and T. H. Watts. 1990. MHC class II-restricted presentation of native protein antigen by B cells is inhibitable by cycloheximide and brefeldin A. J. Immunol.145:812-818.
    OpenUrlAbstract/FREE Full Text
  82. 82.↵
    Strawbridge, A. B., and J. S. Blum. 2007. Autophagy in MHC class II antigen processing. Curr. Opin. Immunol.19:87-92.
    OpenUrlCrossRefPubMed
  83. 83.↵
    Tewari, M. K., G. Sinnathamby, D. Rajagopal, and L. C. Eisenlohr. 2005. A cytosolic pathway for MHC class II-restricted antigen processing that is proteasome and TAP dependent. Nat. Immunol.6:287-294.
    OpenUrlCrossRefPubMedWeb of Science
  84. 84.↵
    Tukey, J. 1977. Exploratory data analysis. Addison-Wesley, Reading, MA.
  85. 85.↵
    Viboud, G. I., and J. B. Bliska. 2005. Yersinia outer proteins: role in modulation of host cell signaling responses and pathogenesis. Annu. Rev. Microbiol.59:69-89.
    OpenUrlCrossRefPubMedWeb of Science
  86. 86.↵
    Vogel, U., I. B. Autenrieth, R. Berner, and J. Heesemann. 1993. Role of plasmid-encoded antigens of Yersinia enterocolitica in humoral immunity against secondary Y. enterocolitica infection in mice. Microb. Pathog.15:23-36.
    OpenUrlCrossRefPubMed
  87. 87.↵
    Von Pawel-Rammingen, U., M. V. Telepnev, G. Schmidt, K. Aktories, H. Wolf-Watz, and R. Rosqvist. 2000. GAP activity of the Yersinia YopE cytotoxin specifically targets the Rho pathway: a mechanism for disruption of actin microfilament structure. Mol. Microbiol.36:737-748.
    OpenUrlCrossRefPubMedWeb of Science
  88. 88.↵
    Wang, R. F., X. Wang, A. C. Atwood, S. L. Topalian, and S. A. Rosenberg. 1999. Cloning genes encoding MHC class II-restricted antigens: mutated CDC27 as a tumor antigen. Science284:1351-1354.
    OpenUrlAbstract/FREE Full Text
  89. 89.↵
    York, I. A., N. Bhutani, S. Zendzian, A. L. Goldberg, and K. L. Rock. 2006. Tripeptidyl peptidase II is the major peptidase needed to trim long antigenic precursors, but is not required for most MHC class I antigen presentation. J. Immunol.177:1434-1443.
    OpenUrlAbstract/FREE Full Text
  90. 90.↵
    York, I. A., A. L. Goldberg, X. Y. Mo, and K. L. Rock. 1999. Proteolysis and class I major histocompatibility complex antigen presentation. Immunol. Rev.172:49-66.
    OpenUrlCrossRefPubMedWeb of Science
  91. 91.↵
    Zhou, D., P. Li, Y. Lin, J. M. Lott, A. D. Hislop, D. H. Canaday, R. R. Brutkiewicz, and J. S. Blum. 2005. Lamp-2a facilitates MHC class II presentation of cytoplasmic antigens. Immunity22:571-581.
    OpenUrlCrossRefPubMedWeb of Science
View Abstract
PreviousNext
Back to top
Download PDF
Citation Tools
Alternative Endogenous Protein Processing via an Autophagy-Dependent Pathway Compensates for Yersinia-Mediated Inhibition of Endosomal Major Histocompatibility Complex Class II Antigen Presentation
Holger Rüssmann, Klaus Panthel, Brigitte Köhn, Stefan Jellbauer, Sebastian E. Winter, Sara Garbom, Hans Wolf-Watz, Sigrid Hoffmann, Silke Grauling-Halama, Gernot Geginat
Infection and Immunity Nov 2010, 78 (12) 5138-5150; DOI: 10.1128/IAI.00155-10

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Infection and Immunity article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Alternative Endogenous Protein Processing via an Autophagy-Dependent Pathway Compensates for Yersinia-Mediated Inhibition of Endosomal Major Histocompatibility Complex Class II Antigen Presentation
(Your Name) has forwarded a page to you from Infection and Immunity
(Your Name) thought you would be interested in this article in Infection and Immunity.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Alternative Endogenous Protein Processing via an Autophagy-Dependent Pathway Compensates for Yersinia-Mediated Inhibition of Endosomal Major Histocompatibility Complex Class II Antigen Presentation
Holger Rüssmann, Klaus Panthel, Brigitte Köhn, Stefan Jellbauer, Sebastian E. Winter, Sara Garbom, Hans Wolf-Watz, Sigrid Hoffmann, Silke Grauling-Halama, Gernot Geginat
Infection and Immunity Nov 2010, 78 (12) 5138-5150; DOI: 10.1128/IAI.00155-10
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

antigen presentation
autophagy
endosomes
Yersinia pseudotuberculosis
Yersinia pseudotuberculosis Infections

Related Articles

Cited By...

About

  • About IAI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #IAIjournal

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0019-9567; Online ISSN: 1098-5522