Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Proc Natl Acad Sci U S A ; 114(29): E5910-E5919, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28679634

ABSTRACT

Although antigen cross-presentation in dendritic cells (DCs) is critical to the initiation of most cytotoxic immune responses, the intracellular mechanisms and traffic pathways involved are still unclear. One of the most critical steps in this process, the export of internalized antigen to the cytosol, has been suggested to be mediated by Sec61. Sec61 is the channel that translocates signal peptide-bearing nascent polypeptides into the endoplasmic reticulum (ER), and it was also proposed to mediate protein retrotranslocation during ER-associated degradation (a process called ERAD). Here, we used a newly identified Sec61 blocker, mycolactone, to analyze Sec61's contribution to antigen cross-presentation, ERAD, and transport of internalized antigens into the cytosol. As shown previously in other cell types, mycolactone prevented protein import into the ER of DCs. Mycolactone-mediated Sec61 blockade also potently suppressed both antigen cross-presentation and direct presentation of synthetic peptides to CD8+ T cells. In contrast, it did not affect protein export from the ER lumen or from endosomes into the cytosol, suggesting that the inhibition of cross-presentation was not related to either of these trafficking pathways. Proteomic profiling of mycolactone-exposed DCs showed that expression of mediators of antigen presentation, including MHC class I and ß2 microglobulin, were highly susceptible to mycolactone treatment, indicating that Sec61 blockade affects antigen cross-presentation indirectly. Together, our data suggest that the defective translocation and subsequent degradation of Sec61 substrates is the cause of altered antigen cross-presentation in Sec61-blocked DCs.


Subject(s)
Antigen Presentation/physiology , Endosomes/metabolism , Macrolides/pharmacology , SEC Translocation Channels/metabolism , Animals , Antigen Presentation/drug effects , Cell Line , Cytosol/drug effects , Cytosol/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , Endosomes/drug effects , HEK293 Cells , Humans , Mice , Protein Transport/drug effects , SEC Translocation Channels/antagonists & inhibitors
2.
Proc Natl Acad Sci U S A ; 110(9): 3393-8, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23401531

ABSTRACT

Endoplasmic reticulum-associated degradation (ERAD) is a constitutive process that identifies misfolded proteins in the ER and shuttles them to the cytosol, where they can be degraded by the proteasome. The accumulation of misfolded proteins can be catastrophic at both the cellular and organismal level. Although the players involved and mechanistic details of ERAD are being characterized, much remains to be learned. Because of the complexity of the pathway, experimental studies generally require labor-intensive biochemical techniques. Here, we report the development of a system to analyze ERAD based on mutants of split or intact Venus fluorescent protein for which fluorescence depends on enzymatic deglycosylation. We have generated variants that only become fluorescent when they are first glycosylated in the ER and subsequently deglycosylated after retrotranslocation into the cytosol. The E3 ubiquitin ligase HMG-coA reductase degradation 1 homolog (Hrd1) and, consistent with the demonstrated glycosylation/deglycosylation requirement, the cytosolic deglycosylating enzyme peptide:N'glycanase are both required for fluorescence. Furthermore, although these deglycosylation-dependent fluorescent proteins are themselves ERAD substrates, they can also be fused to additional ERAD substrates to interrogate substrate-specific pathways. To validate the system we performed a genomewide siRNA screen that successfully identified known ERAD factors such as Hrd1; homocysteine-inducible, endoplasmic reticulum stress-inducible, ubiquitin-like domain member 1 (HERP); sel-1 suppressor of lin-12-like (SEL1L); and p97. These tools should greatly facilitate the identification of ERAD components and investigation of the mechanisms involved in this critical pathway.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Luminescent Proteins/metabolism , Bacterial Proteins/metabolism , Fluorescence , Genome, Human/genetics , Glycosylation , HEK293 Cells , Humans , Mutation/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
Glycobiology ; 25(8): 836-44, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25900930

ABSTRACT

N-Glycanase 1, encoded by NGLY1, catalyzes the deglycosylation of misfolded N-linked glycoproteins retrotranslocated into the cytosol. We identified nine cases with mutations in NGLY1. The patients show developmental delay, seizures, peripheral neuropathy, abnormal liver function and alacrima (absence of tears). The mutations in NGLY1 resulted in the absence of N-glycanase 1 protein in patient-derived fibroblasts. Applying a recently established cellular deglycosylation-dependent Venus fluorescence assay, we found that patient fibroblasts had dramatically reduced fluorescence, indicating a pronounced reduction in N-glycanase enzymatic activity. Using this assay, we could find no evidence of other related activities. Our findings reveal that NGLY1 mutations destroy both N-glycanase 1 protein and enzymatic activity.


Subject(s)
Developmental Disabilities/genetics , Eye Diseases, Hereditary/genetics , Hepatic Insufficiency/genetics , Lacrimal Apparatus Diseases/genetics , Mutation , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Peripheral Nervous System Diseases/genetics , Seizures/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Developmental Disabilities/pathology , Enzyme Assays , Exons , Fibroblasts/enzymology , Fibroblasts/pathology , Gene Expression , Genes, Reporter , Hepatic Insufficiency/congenital , Humans , Lacrimal Apparatus Diseases/congenital , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Peripheral Nervous System Diseases/congenital , Primary Cell Culture , Seizures/congenital
4.
J Immunol ; 185(7): 4336-43, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20802151

ABSTRACT

Exposure to Mycobacterium tuberculosis can result in lifelong but asymptomatic infection in most individuals. Although CD8(+) T cells are elicited at high frequencies over the course of infection in both humans and mice, how phagosomal M. tuberculosis Ags are processed and presented by MHC class I molecules is poorly understood. Broadly, both cytosolic and noncytosolic pathways have been described. We have previously characterized the presentation of three HLA-I epitopes from M. tuberculosis and shown that these Ags are processed in the cytosol, whereas others have demonstrated noncytosolic presentation of the 19-kDa lipoprotein as well as apoptotic bodies from M. tuberculosis-infected cells. In this paper, we now characterize the processing pathway in an additional six M. tuberculosis epitopes from four proteins in human dendritic cells. Addition of the endoplasmic reticulum-Golgi trafficking inhibitor, brefeldin A, resulted in complete abrogation of Ag processing consistent with cytosolic presentation. However, although addition of the proteasome inhibitor epoxomicin blocked the presentation of two epitopes, presentation of four epitopes was enhanced. To further examine the requirement for proteasomal processing of an epoxomicin-enhanced epitope, an in vitro proteasome digestion assay was established. We find that the proteasome does indeed generate the epitope and that epitope generation is enhanced in the presence of epoxomicin. To further confirm that both the epoxomicin-inhibited and epoxomicin-enhanced epitopes are processed cytosolically, we demonstrate that TAP transport and new protein synthesis are required for presentation. Taken together, these data demonstrate that immunodominant M. tuberculosis CD8(+) Ags are processed and presented using a cytosolic pathway.


Subject(s)
Antigen Presentation/immunology , Antigens, Bacterial/immunology , Cytosol/immunology , Immunodominant Epitopes/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytosol/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Epitopes, T-Lymphocyte/immunology , Golgi Apparatus/immunology , Golgi Apparatus/metabolism , Humans , Immunodominant Epitopes/metabolism , Lymphocyte Activation/immunology , Mice , Mycobacterium tuberculosis/immunology , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism
5.
FEBS J ; 289(11): 3115-3131, 2022 06.
Article in English | MEDLINE | ID: mdl-34995415

ABSTRACT

The polycaspase inhibitor Z-VAD-fmk acts as an inhibitor of peptide: N-glycanase (NGLY1), an endoglycosidase which cleaves N-linked glycans from glycoproteins exported from the endoplasmic reticulum (ER) during ER-associated degradation (ERAD). Both pharmacological N-glycanase inhibition by Z-VAD-fmk and siRNA-mediated knockdown (KD) of NGLY1 induce GFP-LC3-positive puncta in HEK 293 cells. The activation of ER stress markers or induction of reactive oxygen species (ROS) is not observed under either condition. Moreover, Ca2+ handling is unaffected when observing release from intracellular stores. Under conditions of pharmacological NGLY1 inhibition or NGLY1 KD, upregulation of autophagosome formation without impairment of autophagic flux is observed. Enrichment of autophagosomes by immunoprecipitation (IP) and mass spectrometry-based proteomic analysis reveals comparable autophagosomal protein content. Gene ontology analysis of proteins enriched in autophagosome IPs shows overrepresentation of factors involved in protein translation, localization and targeting, RNA degradation and protein complex disassembly. Upregulation of autophagy represents a cellular adaptation to NGLY1 inhibition or KD, and ATG13-deficient mouse embryonic fibroblasts (MEFs) show reduced viability under these conditions. In contrast, treatment with pan-caspase inhibitor, Q-VD-OPh, does not induce cellular autophagy. Therefore, experiments with Z-VAD-fmk are complicated by the effects of NGLY1 inhibition, including induction of autophagy, and Q-VD-OPh represents an alternative caspase inhibitor free from this limitation. ENZYMES: Peptide:N-glycanase1, Peptide-N(4)-(N-acetyl-beta-glucosaminyl)asparagine amidase [EC:3.5.1.52].


Subject(s)
Fibroblasts , Proteomics , Animals , Autophagy , Caspases , Fibroblasts/metabolism , HEK293 Cells , Humans , Mice , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/chemistry , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Peptides/metabolism
6.
PLoS Pathog ; 5(4): e1000374, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19360129

ABSTRACT

Mycobacterium tuberculosis (Mtb) resides in a long-lived phagosomal compartment that resists maturation. The manner by which Mtb antigens are processed and presented on MHC Class I molecules is poorly understood. Using human dendritic cells and IFN-gamma release by CD8(+) T cell clones, we examined the processing and presentation pathway for two Mtb-derived antigens, each presented by a distinct HLA-I allele (HLA-Ia versus HLA-Ib). Presentation of both antigens is blocked by the retrotranslocation inhibitor exotoxin A. Inhibitor studies demonstrate that, after reaching the cytosol, both antigens require proteasomal degradation and TAP transport, but differ in the requirement for ER-golgi egress and new protein synthesis. Specifically, presentation by HLA-B8 but not HLA-E requires newly synthesized HLA-I and transport through the ER-golgi. Phenotypic analysis of the Mtb phagosome by flow organellometry revealed the presence of Class I and loading accessory molecules, including TAP and PDI. Furthermore, loaded HLA-I:peptide complexes are present within the Mtb phagosome, with a pronounced bias towards HLA-E:peptide complexes. In addition, protein analysis also reveals that HLA-E is enriched within the Mtb phagosome compared to HLA-A2. Together, these data suggest that the phagosome, through acquisition of ER-localized machinery and as a site of HLA-I loading, plays a vital role in the presentation of Mtb-derived antigens, similar to that described for presentation of latex bead-associated antigens. This is, to our knowledge, the first description of this presentation pathway for an intracellular pathogen. Moreover, these data suggest that HLA-E may play a unique role in the presentation of phagosomal antigens.


Subject(s)
Antigen Presentation/physiology , Antigens, Bacterial/metabolism , Histocompatibility Antigens Class I/metabolism , Mycobacterium tuberculosis/immunology , Phagosomes/metabolism , Antigens, Bacterial/immunology , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Mycobacterium tuberculosis/metabolism , Phagosomes/immunology
7.
J Exp Med ; 196(11): 1473-81, 2002 Dec 02.
Article in English | MEDLINE | ID: mdl-12461082

ABSTRACT

Previous studies in mice and humans have suggested an important role for CD8+ T cells in host defense to Mtb. Recently, we have described human, Mtb-specific CD8+ cells that are neither HLA-A, B, or C nor group 1 CD1 restricted, and have found that these cells comprise the dominant CD8+ T cell response in latently infected individuals. In this report, three independent methods are used to demonstrate the ability of these cells to recognize Mtb-derived antigen in the context of the monomorphic HLA-E molecule. This is the first demonstration of the ability of HLA-E to present pathogen-derived antigen. Further definition of the HLA-E specific response may aid development of an effective vaccine against tuberculosis.


Subject(s)
Antigen Presentation , Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , HLA Antigens/physiology , Histocompatibility Antigens Class I/physiology , Mycobacterium tuberculosis/immunology , Antigens, CD/physiology , Cell Line , Dendritic Cells/physiology , Humans , Interferon-gamma/biosynthesis , Lectins, C-Type/physiology , NK Cell Lectin-Like Receptor Subfamily D , Receptors, Immunologic/physiology , Receptors, Natural Killer Cell , HLA-E Antigens
8.
Blood Adv ; 3(22): 3661-3673, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31751472

ABSTRACT

Acute myeloid leukemias (AML) harboring a constitutively active internal tandem duplication (ITD) mutation in the FMS-like kinase tyrosine kinase (FLT3) receptor are associated with poor patient prognosis. Despite initial clinical responses to FLT3 kinase inhibitors, patients eventually relapse. Mechanisms of resistance include the acquisition of secondary FLT3 mutations and protective stromal signaling within the bone marrow niche. Here we show that LAM-003, a prodrug of the heat shock protein 90 inhibitor LAM-003A, has cytotoxic activity against AML cell lines and primary samples harboring FLT3-ITD. LAM-003 regressed tumors in an MV-4-11 xenograft mouse model and extended survival in a MOLM-13 systemic model. LAM-003 displayed synergistic activity with chemotherapeutic drugs and FLT3 inhibitors, with the most robust synergy being obtained with venetoclax, a BCL-2 inhibitor. This finding was verified in a MOLM-13 systemic survival model in which the combination significantly prolonged survival compared with the single agents. Importantly, LAM-003 exhibited equipotent activity against FLT3 inhibitor-resistant mutants of FLT3, such as D835 or F691, in cytotoxic and FLT3 degradation assays. LAM-003 also retained potency in AML cells grown in stromal-conditioned media that were resistant to FLT3 inhibitors. Lastly, a genome-wide CRISPR screen revealed epigenetic regulators, including KDM6A, as determinants of LAM-003 sensitivity in AML cell lines, leading to the discovery of synergy with an EZH2 inhibitor. Collectively, these preclinical findings support the use of LAM-003 in FLT3-ITD patients with AML who no longer respond to FLT3 inhibitor therapy either as a single agent or in combination with drugs known to be active in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Duplication , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Epigenesis, Genetic , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Mice , Mutation , Protein Kinase Inhibitors/therapeutic use
9.
Nat Commun ; 9(1): 3104, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082832

ABSTRACT

Dendritic cells use a specialized pathway called cross-presentation to activate CD8+ T cells by presenting peptides from exogenous protein antigens on major histocompatibility complex class I molecules. Considerable evidence suggests that internalized antigens cross endocytic membranes to access cytosolic proteasomes for processing. The mechanism of protein dislocation represents a major unsolved problem. Here we describe the development of a sensitive reporter substrate, an N-glycosylated variant of Renilla luciferase fused to the Fc region of human IgG1. The luciferase variant is designed to be enzymatically inactive when glycosylated, but active after the asparagine to aspartic acid conversion that occurs upon deglycosylation by the cytosolic enzyme N-glycanase-1. The generation of cytosolic luminescence depends on internalization, deglycosylation, the cytosolic AAA-ATPase VCP/p97, and the cytosolic chaperone HSP90. By incorporating a T cell epitope into the fusion protein, we demonstrate that antigen dislocation into the cytosol is the rate limiting step in cross-presentation.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cross-Priming , Cytosol/metabolism , Immunoglobulin G/metabolism , Animals , Antigen Presentation , Antigens/metabolism , Dendritic Cells/immunology , Endocytosis , Epitopes/chemistry , Glycosylation , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , Histocompatibility Antigens Class I/immunology , Humans , Luciferases/metabolism , Molecular Chaperones/metabolism , Protein Binding , Protein Transport , Renilla
10.
Nat Commun ; 9(1): 5110, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504895

ABSTRACT

Non-small cell lung carcinoma (NSCLC) is leading cause of cancer-related deaths in the world. The Tumor Suppressor Candidate 3 (TUSC3) at chromosome 8p22 known to be frequently deleted in cancer is often found to be deleted in advanced stage of solid tumors. However, the role of TUSC3 still remains controversial in lung cancer and context-dependent in several cancers. Here we propose that miR-224/-520c-dependent TUSC3 deficiency enhances the metastatic potential of NSCLC through the alteration of three unfolded protein response pathways and HRD1-dependent ERAD. ATF6α-dependent UPR is enhanced whereas the affinity of HRD1 to its substrates, PERK, IRE1α and p53 is weakened. Consequently, the alteration of UPRs and the suppressed p53-NM23H1/2 pathway by TUSC3 deficiency is ultimately responsible for enhancing metastatic potential of lung cancer. These findings provide mechanistic insight of unrecognized roles of TUSC3 in cancer progression and the oncogenic role of HRD1-dependent ERAD in cancer metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Endoplasmic Reticulum-Associated Degradation/physiology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/metabolism , MicroRNAs/metabolism , Tumor Suppressor Proteins/metabolism , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Endoplasmic Reticulum-Associated Degradation/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , In Situ Hybridization , Lung Neoplasms/genetics , Membrane Proteins/genetics , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Xenograft Model Antitumor Assays
11.
Curr Opin Immunol ; 46: 89-96, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28528219

ABSTRACT

Cross-presentation is an MHC-I antigen processing pathway that results in the presentation of peptides from exogenous viral, bacterial, parasitic, and tumor antigens and ultimately leads to priming of naïve CD8+ T cells. This process involves several cellular compartments and multiple components. Successful generation of MHC-I-peptide complexes requires that these components act together in a coordinated fashion. We discuss recent findings on the source of MHC-I, the role of the TAP transporter, the importance of intracellular trafficking events, mechanisms of antigen access the cytosol, and how innate immune signals can affect presentation, with an emphasis on how these pathways compare to conventional antigen presentation and how they correlate with existing data.


Subject(s)
Antigen Presentation/immunology , Cross-Priming/immunology , Histocompatibility Antigens Class I/immunology , Animals , Antigens/immunology , Antigens/metabolism , Biological Transport , Cytosol/immunology , Cytosol/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Peptides/immunology , Peptides/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
12.
Microbes Infect ; 8(11): 2587-98, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16952476

ABSTRACT

Mycobacterium tuberculosis infection in non-human primates parallels human tuberculosis, and provides a valuable vaccine evaluation model. However, this model is limited by the availability of real-time, non-invasive information regarding disease progression. Consequently, we have combined computed tomography scanning with enumeration of antigen-specific T cell responses. Four rhesus monkeys were infected with M. tuberculosis strain H37Rv (1000 cfu) in the right lower lobe via a bronchoscope. All uniformly developed progressive tuberculosis, and required euthanasia at 12 weeks. Computed tomography scanning provided detailed real-time imaging of disease progression. At necropsy, computed tomography and pathohistologic findings were tightly correlated, and characteristic of human disease. Immunologic monitoring demonstrated progressive evolution of high frequency M. tuberculosis-specific CD4(+) and CD8(+) T cell responses. Peripheral blood effector cell frequencies were similar to those observed in tissues. In summary, computed tomography scanning in conjunction with immunologic monitoring provides a non-invasive, accurate, and rapid assessment of tuberculosis in the non-human primate.


Subject(s)
Disease Models, Animal , Lung/diagnostic imaging , Macaca mulatta , Tuberculosis/diagnostic imaging , Tuberculosis/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Progression , Histocytochemistry , Lung/microbiology , Lung/pathology , Mycobacterium tuberculosis/isolation & purification , Tomography, X-Ray Computed , Tuberculosis/microbiology , Tuberculosis/pathology
13.
Microbes Infect ; 7(4): 776-88, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15823514

ABSTRACT

Tuberculosis remains a global health concern. Control of infection is dependent on cell-mediated immune responses, with CD4+ T lymphocytes playing a central role. In this article, data supporting the importance of CD8+ T lymphocytes is reviewed, with an emphasis on the unique functional roles that this lymphocyte subset may play.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/prevention & control , Animals , Antigen Presentation , Humans , Immunologic Surveillance , Mice , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology
14.
Mol Immunol ; 68(2 Pt A): 112-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26005101

ABSTRACT

A long unanswered question in the antigen presentation field is how exogenous antigens cross-presented by Major Histocompatibility Complex class I (MHC-I) molecules to CD8(+) T cells are translocated into the cytosol. Here we discuss the known mechanisms involved in this process with a focus on the hypothesized role of the machinery that functions in endoplasmic reticulum-associated degradation (ERAD). Other potential mechanisms of antigen entry to the cytosol are also discussed.


Subject(s)
Antigen Presentation , Antigens/immunology , Dendritic Cells/immunology , Endoplasmic Reticulum-Associated Degradation/immunology , Histocompatibility Antigens Class I/immunology , Animals , Antigens/genetics , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Cytosol/immunology , Cytosol/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation/genetics , Histocompatibility Antigens Class I/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Protein Transport , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
15.
PLoS One ; 8(1): e51343, 2013.
Article in English | MEDLINE | ID: mdl-23326313

ABSTRACT

Gamma-interferon-inducible lysosomal thiol reductase (GILT) is known to reduce disulfide bonds present in proteins internalized by antigen presenting cells, facilitating optimal processing and presentation of peptides on Major Histocompatibility Complex class II molecules, as well as the subsequent activation of CD4-positive T lymphocytes. Here, we show that GILT is required for class II-restricted processing and presentation of immunodominant epitopes from the major house dust mite allergen Der p 1. In the absence of GILT, CD4-positive T cell responses to Der p 1 are significantly reduced, resulting in mitigated allergic airway inflammation in response to Der p 1 and house dust mite extracts in a murine model of asthma.


Subject(s)
Antigens, Dermatophagoides/immunology , Arthropod Proteins/immunology , Cysteine Endopeptidases/immunology , Histocompatibility Antigens Class II/immunology , Oxidoreductases/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Aspartic Acid Endopeptidases/immunology , Asthma/immunology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Cockroaches/immunology , Coculture Techniques , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases/genetics , Oxidoreductases/metabolism , Oxidoreductases Acting on Sulfur Group Donors , Pyroglyphidae/immunology , Reverse Transcriptase Polymerase Chain Reaction
16.
PLoS One ; 4(3): e4807, 2009.
Article in English | MEDLINE | ID: mdl-19277197

ABSTRACT

BACKGROUND: Dendritic cells (DC) are the most potent antigen presenting cells (APC) of the immune system. Prostaglandin E(2), cyclic AMP, and protein kinase A (PKA) have all been shown to regulate DC maturation and activity. In other cells, the ability of these molecules to convey their signals has been shown to be dependent on A-kinase anchoring proteins (AKAPs). Here we present evidence for the existence and functional importance of AKAPs in human DC. METHODOLOGY/PRINCIPAL FINDINGS: Using immunofluorescence and/or western analyses we identify AKAP79, AKAP149, AKAP95, AKAP LBC and Ezrin. We also demonstrate by western analysis that expression of AKAP79, AKAP149 and RII are upregulated with DC differentiation and maturation. We establish the functional importance of PKA anchoring in multiple aspects of DC biology using the anchoring inhibitor peptides Ht31 and AKAP-IS. Incubation of protein or peptide antigen loaded DC with Ht31 or AKAP-IS results in a 30-50% decrease in antigen presentation as measured by IFN-gamma production from antigen specific CD4(+) T cells. Incubation of LPS treated DC with Ht31 results in 80% inhibition of TNF-alpha and IL-10 production. Ht31 slightly decreases the expression of CD18 and CD11a and CD11b, slightly increases the basal expression of CD83, dramatically decreases the LPS stimulated expression of CD40, CD80 and CD83, and significantly increases the expression of the chemokine receptor CCR7. CONCLUSIONS: These experiments represent the first evidence for the functional importance of PKA anchoring in multiple aspects of DC biology.


Subject(s)
A Kinase Anchor Proteins/physiology , Antigen Presentation/physiology , Dendritic Cells/immunology , A Kinase Anchor Proteins/antagonists & inhibitors , Amino Acid Sequence , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dinoprostone/pharmacology , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/pharmacology , Molecular Sequence Data , Monocytes/cytology , Monocytes/drug effects , Proteins/pharmacology , Receptors, CCR7/biosynthesis , Receptors, CCR7/genetics , Subcellular Fractions/chemistry , Tumor Necrosis Factor-alpha/metabolism
17.
J Immunol ; 177(1): 437-42, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785540

ABSTRACT

CD8+ T cells play an important role in the host response to infection with Mycobacterium tuberculosis (Mtb). Mtb resides in an arrested phagosome that is phenotypically similar to an early endosome. The mechanisms by which Mtb-derived Ags gain access to the HLA-I-processing pathway are incompletely characterized. Studies with CD8+ T cell lines have suggested that Mtb Ags gain access to the HLA-I pathway in an alternate vacuolar pathway that is both brefeldin A (BFA) and TAP independent. To define the requirements of entry of Ag into the HLA-I pathway, we have used human CD8+ T cell clones specific for the secreted Mtb Ag CFP10. Human monocyte-derived dendritic cells were pulsed with CFP10 expressed in a recombinant adenovirus, surface adsorbed to microspheres, or in its native form by Mtb. When delivered by adenovirus, processing and presentation of CFP10 were blocked by both BFA and the proteasomal blocker lactacystin. In contrast, processing of CFP10 adsorbed to the surface of microspheres was not affected by either of these Ag-processing inhibitors. BFA, lactacystin, and TAP inhibition blocked the recognition of Mtb-infected dendritic cells, suggesting that processing was via a cytosolic pathway for this secreted protein Ag. We conclude that secreted proteins from Mtb can be processed in a BFA- and proteasome-dependent manner, consistent with egress of Ag into the cytosol and subsequent loading of proteasomally derived peptides.


Subject(s)
Antigen Presentation/immunology , Bacterial Proteins/metabolism , Cytosol/metabolism , Cytosol/microbiology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Mycobacterium tuberculosis/immunology , Signal Transduction/immunology , ATP-Binding Cassette Transporters/antagonists & inhibitors , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Amino Acid Sequence , Antigen Presentation/drug effects , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Brefeldin A/pharmacology , Cell Line , Cell Line, Transformed , Clone Cells , Cytosol/immunology , Humans , Immediate-Early Proteins/physiology , Lactic Acid/immunology , Lactic Acid/metabolism , Leupeptins/pharmacology , Microspheres , Molecular Sequence Data , Mycobacterium tuberculosis/pathogenicity , Polyglycolic Acid/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/metabolism , Proteasome Inhibitors , Signal Transduction/drug effects , Viral Proteins/physiology
18.
Semin Respir Infect ; 18(4): 320-38, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14679479

ABSTRACT

Tuberculosis (TB) remains a leading cause of infectious disease mortality worldwide, accounting for nearly 2 million deaths annually. Despite the availability of effective anti-TB therapy, the world's case burden of TB continues to climb, in part owing to the concurrent acquired immune deficiency syndrome pandemic. The widespread use of the current TB vaccine, the bacille Calmette-Guérin strain of M. bovis (BCG), has failed to curtail the TB epidemic and therefore strategies for the eradication of TB have centered on aggressive case-finding and managed treatment, such as directly observed treatment short course (DOTS). Although DOTS continues to have a significant impact, the ability of DOTS to eradicate TB is limited. Therefore, TB eradication will require the development of an improved vaccine, which, in turn, will require application of state-of-the-art vaccine technology and of our knowledge of TB immunology and TB genomics to this problem. In this article, knowledge about the requisite components of protective immunity, modern vaccine strategies, and elements of clinical trials required to evaluate vaccine efficacy, as well as the status of current TB vaccine candidates, are reviewed.


Subject(s)
BCG Vaccine/administration & dosage , Global Health , Immunization Programs/organization & administration , Infection Control/standards , Mycobacterium tuberculosis/immunology , Tuberculosis/prevention & control , Female , Forecasting , Humans , Incidence , Infection Control/trends , Male , Mass Screening/standards , Mass Screening/trends , Primary Prevention/methods , Risk Assessment , Sensitivity and Specificity , Tuberculosis/epidemiology
19.
Mol Microbiol ; 51(2): 359-70, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14756778

ABSTRACT

The RD1 genomic region is present in virulent strains of Mycobacterium tuberculosis (MTB), missing from the vaccine strain M. bovis BCG, and its importance to virulence has been established experimentally. Based on in silico analysis, it has been suggested that RD1 may encode a novel secretion system, but the mechanism by which this region affects virulence is unknown. Here we examined mutants disrupted in five individual RD1 genes. Both in vitro and in vivo, each mutant displayed an attenuated phenotype very similar to a mutant missing the entire RD1 region. Genetic complementation of individual genes restored virulence. Attenuated mutants could multiply within THP-1 cells, but they were unable to spread to uninfected macrophages. We also examined export of two immunodominant RD1 proteins, CFP-10 and ESAT-6. Export of these proteins was greatly reduced or abolished in each attenuated mutant. Again, genetic complementation restored a wild-type phenotype. Our results indicate that RD1 genes work together to form a single virulence determinant, and argue that RD1 encodes a novel specialized secretion system that is required for pathogenesis of MTB.


Subject(s)
Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Animals , BCG Vaccine/genetics , Base Sequence , Cell Line , Chromosome Mapping , DNA Primers , Genes, Bacterial/genetics , Genetic Complementation Test , Genetic Vectors , Humans , Mice , Mice, Inbred C57BL , Mutagenesis , Phenotype , Sequence Deletion , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL