1
|
Chen J, Wang X, Schmalen A, Haines S, Wolff M, Ma H, Zhang H, Stoleriu MG, Nowak J, Nakayama M, Bueno M, Brands J, Mora AL, Lee JS, Krauss-Etschmann S, Dmitrieva A, Frankenberger M, Hofer TP, Noessner E, Moosmann A, Behr J, Milger K, Deeg CA, Staab-Weijnitz CA, Hauck SM, Adler H, Goldmann T, Gaede KI, Behrends J, Kammerl IE, Meiners S. Antiviral CD8 + T-cell immune responses are impaired by cigarette smoke and in COPD. Eur Respir J 2023; 62:2201374. [PMID: 37385655 PMCID: PMC10397470 DOI: 10.1183/13993003.01374-2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 05/24/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Virus infections drive COPD exacerbations and progression. Antiviral immunity centres on the activation of virus-specific CD8+ T-cells by viral epitopes presented on major histocompatibility complex (MHC) class I molecules of infected cells. These epitopes are generated by the immunoproteasome, a specialised intracellular protein degradation machine, which is induced by antiviral cytokines in infected cells. METHODS We analysed the effects of cigarette smoke on cytokine- and virus-mediated induction of the immunoproteasome in vitro, ex vivo and in vivo using RNA and Western blot analyses. CD8+ T-cell activation was determined in co-culture assays with cigarette smoke-exposed influenza A virus (IAV)-infected cells. Mass-spectrometry-based analysis of MHC class I-bound peptides uncovered the effects of cigarette smoke on inflammatory antigen presentation in lung cells. IAV-specific CD8+ T-cell numbers were determined in patients' peripheral blood using tetramer technology. RESULTS Cigarette smoke impaired the induction of the immunoproteasome by cytokine signalling and viral infection in lung cells in vitro, ex vivo and in vivo. In addition, cigarette smoke altered the peptide repertoire of antigens presented on MHC class I molecules under inflammatory conditions. Importantly, MHC class I-mediated activation of IAV-specific CD8+ T-cells was dampened by cigarette smoke. COPD patients exhibited reduced numbers of circulating IAV-specific CD8+ T-cells compared to healthy controls and asthmatics. CONCLUSION Our data indicate that cigarette smoke interferes with MHC class I antigen generation and presentation and thereby contributes to impaired activation of CD8+ T-cells upon virus infection. This adds important mechanistic insight on how cigarette smoke mediates increased susceptibility of smokers and COPD patients to viral infections.
Collapse
Affiliation(s)
- Jie Chen
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
- These authors contributed equally
| | - Xinyuan Wang
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Guangzhou Medical University, Guangzhou, China
- These authors contributed equally
| | - Adrian Schmalen
- Department of Veterinary Sciences, LMU Munich, Martinsried, Germany
- Metabolomics and Proteomics Core, Helmholtz Center Munich, Munich, Germany
| | - Sophia Haines
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Martin Wolff
- Institute of Experimental Medicine, Christian-Albrechts University Kiel, Kiel, Germany
| | - Huan Ma
- Institute of Experimental Medicine, Christian-Albrechts University Kiel, Kiel, Germany
| | - Huabin Zhang
- Neurosurgery Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mircea Gabriel Stoleriu
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Division of Thoracic Surgery Munich, University Clinic of Ludwig-Maximilians-University of Munich (LMU), Munich, Germany
- Asklepios Pulmonary Hospital, Gauting, Germany
| | - Johannes Nowak
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Misako Nakayama
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Marta Bueno
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Judith Brands
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ana L Mora
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart Lung Institute, Ohio State University, Columbus, OH, USA
| | - Janet S Lee
- Division of Pulmonary and Critical Care Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Anna Dmitrieva
- Institute of Asthma and Allergy Prevention, Helmholtz Center Munich, Member of the German Center of Lung Research (DZL), Munich, Germany
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Marion Frankenberger
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas P Hofer
- Immunoanalytics - Working Group Tissue Control of Immunocytes, Helmholtz Center Munich, Munich, Germany
| | - Elfriede Noessner
- Immunoanalytics - Working Group Tissue Control of Immunocytes, Helmholtz Center Munich, Munich, Germany
| | - Andreas Moosmann
- DZIF Group Host Control of Viral Latency and Reactivation, Department of Medicine III, LMU-Klinikum, Munich, Germany
- DZIF - German Center for Infection Research, Munich, Germany
| | - Jürgen Behr
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Katrin Milger
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Cornelia A Deeg
- Department of Veterinary Sciences, LMU Munich, Martinsried, Germany
| | - Claudia A Staab-Weijnitz
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Center Munich, Munich, Germany
| | - Heiko Adler
- Institute of Asthma and Allergy Prevention, Helmholtz Center Munich, Member of the German Center of Lung Research (DZL), Munich, Germany
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Torsten Goldmann
- Histology, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Karoline I Gaede
- BioMaterialBank North, Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Popgen 2.0 Network, (P2N), Borstel, Germany
| | - Jochen Behrends
- Core Facility Fluorescence Cytometry, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Ilona E Kammerl
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- These authors contributed equally
| | - Silke Meiners
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Medicine, Christian-Albrechts University Kiel, Kiel, Germany
- Research Center Borstel, Leibniz Lung Center, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
- These authors contributed equally
| |
Collapse
|
2
|
Sarkar S, Mishra A, Periasamy S, Dyett B, Dogra P, Ball AS, Yeo LY, White JF, Wang Z, Cristini V, Jagannath C, Khan A, Soni SK, Drummond CJ, Conn CE. Prospective Subunit Nanovaccine against Mycobacterium tuberculosis Infection─Cubosome Lipid Nanocarriers of Cord Factor, Trehalose 6,6' Dimycolate. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37262346 DOI: 10.1021/acsami.3c04063] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
An improved vaccine is urgently needed to replace the now more than 100-year-old Bacillus Calmette-Guérin (BCG) vaccine against tuberculosis (TB) disease, which represents a significant burden on global public health. Mycolic acid, or cord factor trehalose 6,6' dimycolate (TDM), a lipid component abundant in the cell wall of the pathogen Mycobacterium tuberculosis (MTB), has been shown to have strong immunostimulatory activity but remains underexplored due to its high toxicity and poor solubility. Herein, we employed a novel strategy to encapsulate TDM within a cubosome lipid nanocarrier as a potential subunit nanovaccine candidate against TB. This strategy not only increased the solubility and reduced the toxicity of TDM but also elicited a protective immune response to control MTB growth in macrophages. Both pre-treatment and concurrent treatment of the TDM encapsulated in lipid monoolein (MO) cubosomes (MO-TDM) (1 mol %) induced a strong proinflammatory cytokine response in MTB-infected macrophages, due to epigenetic changes at the promoters of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in comparison to the untreated control. Furthermore, treatment with MO-TDM (1 mol %) cubosomes significantly improved antigen processing and presentation capabilities of MTB-infected macrophages to CD4 T cells. The ability of MO-TDM (1 mol %) cubosomes to induce a robust innate and adaptive response in vitro was further supported by a mathematical modeling study predicting the vaccine efficacy in vivo. Overall, these results indicate a strong immunostimulatory effect of TDM when delivered through the lipid nanocarrier, suggesting its potential as a novel TB vaccine.
Collapse
Affiliation(s)
- Sampa Sarkar
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Selvakannan Periasamy
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Brendan Dyett
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, United States
| | - Andrew S Ball
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Leslie Y Yeo
- School of Engineering, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Jacinta F White
- The Commonwealth Scientific and Industrial Research Organisation, Clayton 3169, Victoria, Australia
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, United States
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Imaging Physics, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, United States
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York 10021, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Sarvesh K Soni
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Charlotte E Conn
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| |
Collapse
|
3
|
Wang X, Zhang H, Wang Y, Bramasole L, Guo K, Mourtada F, Meul T, Hu Q, Viteri V, Kammerl I, Konigshoff M, Lehmann M, Magg T, Hauck F, Fernandez IE, Meiners S. DNA sensing via the cGAS/STING pathway activates the immunoproteasome and adaptive T-cell immunity. EMBO J 2023; 42:e110597. [PMID: 36912165 PMCID: PMC10106989 DOI: 10.15252/embj.2022110597] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/14/2023] Open
Abstract
The immunoproteasome is a specialized type of proteasome involved in MHC class I antigen presentation, antiviral adaptive immunity, autoimmunity, and is also part of a broader response to stress. Whether the immunoproteasome is regulated by DNA stress, however, is not known. We here demonstrate that mitochondrial DNA stress upregulates the immunoproteasome and MHC class I antigen presentation pathway via cGAS/STING/type I interferon signaling resulting in cell autonomous activation of CD8+ T cells. The cGAS/STING-induced adaptive immune response is also observed in response to genomic DNA and is conserved in epithelial and mesenchymal cells of mice and men. In patients with idiopathic pulmonary fibrosis, chronic activation of the cGAS/STING-induced adaptive immune response in aberrant lung epithelial cells concurs with CD8+ T-cell activation in diseased lungs. Genetic depletion of the immunoproteasome and specific immunoproteasome inhibitors counteract DNA stress induced cytotoxic CD8+ T-cell activation. Our data thus unravel cytoplasmic DNA sensing via the cGAS/STING pathway as an activator of the immunoproteasome and CD8+ T cells. This represents a novel potential pathomechanism for pulmonary fibrosis that opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Xinyuan Wang
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Huabin Zhang
- Neurosurgical Research, Department of Neurosurgery, University Hospital and Walter-Brendel-Centre of Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany.,The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqin Wang
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Laylan Bramasole
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Kai Guo
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Fatima Mourtada
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Thomas Meul
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Qianjiang Hu
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Valeria Viteri
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Ilona Kammerl
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Melanie Konigshoff
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany.,Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mareike Lehmann
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Magg
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Isis E Fernandez
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,Department of Medicine V, University Hospital, LMU Munich, Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
4
|
Kwak C, Nguyen QT, Kim J, Kim TH, Poo H. Influenza Chimeric Protein (3M2e-3HA2-NP) Adjuvanted with PGA/Alum Confers Cross-Protection against Heterologous Influenza A Viruses. J Microbiol Biotechnol 2021; 31:304-316. [PMID: 33263336 PMCID: PMC9705887 DOI: 10.4014/jmb.2011.11029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
Vaccination is the most effective way to prevent influenza virus infections. However, conventional vaccines based on hemagglutinin (HA) have to be annually updated because the HA of influenza viruses constantly mutates. In this study, we produced a 3M2e-3HA2-NP chimeric protein as a vaccine antigen candidate using an Escherichia coli expression system. The vaccination of chimeric protein (15 μg) conferred complete protection against A/Puerto Rico/8/1934 (H1N1; PR8) in mice. It strongly induced influenza virus-specific antibody responses, cytotoxic T lymphocyte activity, and antibody-dependent cellular cytotoxicity. To spare the dose and enhance the cross-reactivity of the chimeric, we used a complex of poly-γ-glutamic acid and alum (PGA/alum) as an adjuvant. PGA/alum-adjuvanted, low-dose chimeric protein (1 or 5 μg) exhibited higher cross-protective effects against influenza A viruses (PR8, CA04, and H3N2) compared with those of chimeric alone or alum-adjuvanted proteins in vaccinated mice. Moreover, the depletion of CD4+ T, CD8+ T, and NK cells reduced the survival rate and efficacy of the PGA/alum-adjuvanted chimeric protein. Collectively, the vaccination of PGA/alum-adjuvanted chimeric protein induced strong protection efficacy against homologous and heterologous influenza viruses in mice, which suggests that it may be a promising universal influenza vaccine candidate.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Alum Compounds/administration & dosage
- Animals
- Antibodies, Viral/immunology
- Cross Reactions
- Female
- Hemagglutinins, Viral
- Humans
- Immunity, Humoral
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Nucleocapsid Proteins/administration & dosage
- Nucleocapsid Proteins/genetics
- Nucleocapsid Proteins/immunology
- Polyglutamic Acid/administration & dosage
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Viral Matrix Proteins/administration & dosage
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
Collapse
Affiliation(s)
- Chaewon Kwak
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Quyen Thi Nguyen
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jaemoo Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Tae-Hwan Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
| | - Haryoung Poo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 344, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
5
|
Restrepo BI, Khan A, Singh VK, Erica de-Leon, Aguillón-Durán GP, Ledezma-Campos E, Canaday DH, Jagannath C. Human monocyte-derived macrophage responses to M. tuberculosis differ by the host's tuberculosis, diabetes or obesity status, and are enhanced by rapamycin. Tuberculosis (Edinb) 2021; 126:102047. [PMID: 33418150 PMCID: PMC7887072 DOI: 10.1016/j.tube.2020.102047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/17/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023]
Abstract
Human macrophages play a major role in controlling tuberculosis (TB), but their anti-mycobacterial mechanisms remain unclear among individuals with metabolic alterations like obesity (TB protective) or diabetes (TB risk). To help discern this, we aimed to: i) Evaluate the impact of the host's TB status or their comorbidities on the anti-mycobacterial responses of their monocyte-derived macrophages (MDMs), and ii) determine if the autophagy inducer rapamycin, can enhance these responses. We used MDMs from newly diagnosed TB patients, their close contacts and unexposed controls. The MDMs from TB patients had a reduced capacity to activate T cells (surrogate for antigen presentation) or kill M. tuberculosis (Mtb) when compared to non-TB controls. The MDMs from obese participants had a higher antigen presenting capacity, whereas those from chronic diabetes patients displayed lower Mtb killing. The activation of MDMs with rapamycin led to an enhanced anti-mycobacterial activity irrespective of TB status but was not as effective in patients with diabetes. Further studies are warranted using MDMs from TB patients with or without metabolic comorbidities to: i) elucidate the mechanisms through which host factors affect Mtb responses, and ii) evaluate host directed therapy using autophagy-inducing drugs like rapamycin to enhance macrophage function.
Collapse
Affiliation(s)
- Blanca I Restrepo
- University of Texas Health Houston, School of Public Health, One West University Blvd, Brownsville, TX, USA; University of Texas Rio Grande Valley, School of Medicine, South Texas Diabetes and Obesity Institute, 1214 W Schunior, Edinburg, TX, USA.
| | - Arshad Khan
- Houston Methodist Research Institute, Houston, Weill-Cornell Medicine, TX, USA.
| | - Vipul K Singh
- Houston Methodist Research Institute, Houston, Weill-Cornell Medicine, TX, USA.
| | - Erica de-Leon
- University of Texas Health Houston, School of Public Health, One West University Blvd, Brownsville, TX, USA.
| | - Génesis P Aguillón-Durán
- University of Texas Health Houston, School of Public Health, One West University Blvd, Brownsville, TX, USA; Secretaria de Salud de Tamaulipas, Reynosa and Ciudad Victoria, Mexico.
| | | | - David H Canaday
- Division of Infectious Disease, Case Western Reserve University and Cleveland, VA, OH, USA.
| | | |
Collapse
|
6
|
Fehlings M, Drobbe L, Beigier-Bompadre M, Viveros PR, Moos V, Schneider T, Meyer TF, Aebischer T, Ignatius R. Usage of Murine T-cell Hybridoma Cells as Responder Cells Reveals Interference of Helicobacter Pylori with Human Dendritic Cell-mediated Antigen Presentation. Eur J Microbiol Immunol (Bp) 2016; 6:306-311. [PMID: 27980859 PMCID: PMC5146649 DOI: 10.1556/1886.2016.00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 10/18/2016] [Indexed: 11/23/2022] Open
Abstract
Direct effects of Helicobacter pylori (H. pylori) on human CD4+ T-cells hamper disentangling a possible bacterial-mediated interference with major histocompatibility complex class II (MHC-II)-dependent antigen presentation to these cells. To overcome this limitation, we employed a previously described assay, which enables assessing human antigen-processing cell function by using murine T-cell hybridoma cells restricted by human leukocyte antigen (HLA) alleles. HLA-DR1+ monocyte-derived dendritic cells were exposed to H. pylori and pulsed with the antigen 85B from Mycobacterium tuberculosis (M. tuberculosis). Interleukin-2 (IL-2) secretion by AG85Baa97-112-specific hybridoma cells was then evaluated as an integral reporter of cognate antigen presentation. This methodology enabled revealing of interference of H. pylori with the antigen-presenting capacity of human dendritic cells.
Collapse
Affiliation(s)
- Michael Fehlings
- Department of Molecular Biology, Max Planck Institute for Infection Biology , Charitéplatz 1, 10117 Berlin, Germany
| | - Lea Drobbe
- Department of Molecular Biology, Max Planck Institute for Infection Biology , Charitéplatz 1, 10117 Berlin, Germany
| | - Macarena Beigier-Bompadre
- Department of Molecular Biology, Max Planck Institute for Infection Biology , Charitéplatz 1, 10117 Berlin, Germany
| | - Pablo Renner Viveros
- Institute for Microbiology and Hygiene, Charité - Universitätsmedizin Berlin , 12203 Berlin, Germany
| | - Verena Moos
- Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin , Hindenburgdamm 30, 12200 Berlin, Germany
| | - Thomas Schneider
- Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin , Hindenburgdamm 30, 12200 Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology , Charitéplatz 1, 10117 Berlin, Germany
| | - Toni Aebischer
- Mycotic and Parasitic Agents and Mycobacteria, Department of Infectious Diseases, Robert Koch-Institute , Berlin, Germany
| | - Ralf Ignatius
- Institute for Microbiology and Hygiene, Charité - Universitätsmedizin Berlin , 12203 Berlin, Germany
| |
Collapse
|
7
|
Maggi J, Schinnerling K, Pesce B, Hilkens CM, Catalán D, Aguillón JC. Dexamethasone and Monophosphoryl Lipid A-Modulated Dendritic Cells Promote Antigen-Specific Tolerogenic Properties on Naive and Memory CD4 + T Cells. Front Immunol 2016; 7:359. [PMID: 27698654 PMCID: PMC5027201 DOI: 10.3389/fimmu.2016.00359] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/01/2016] [Indexed: 12/20/2022] Open
Abstract
Tolerogenic dendritic cells (DCs) are a promising tool to control T cell-mediated autoimmunity. Here, we evaluate the ability of dexamethasone-modulated and monophosphoryl lipid A (MPLA)-activated DCs [MPLA-tolerogenic DCs (tDCs)] to exert immunomodulatory effects on naive and memory CD4+ T cells in an antigen-specific manner. For this purpose, MPLA-tDCs were loaded with purified protein derivative (PPD) as antigen and co-cultured with autologous naive or memory CD4+ T cells. Lymphocytes were re-challenged with autologous PPD-pulsed mature DCs (mDCs), evaluating proliferation and cytokine production by flow cytometry. On primed-naive CD4+ T cells, the expression of regulatory T cell markers was evaluated and their suppressive ability was assessed in autologous co-cultures with CD4+ effector T cells and PPD-pulsed mDCs. We detected that memory CD4+ T cells primed by MPLA-tDCs presented reduced proliferation and proinflammatory cytokine expression in response to PPD and were refractory to subsequent stimulation. Naive CD4+ T cells were instructed by MPLA-tDCs to be hyporesponsive to antigen-specific restimulation and to suppress the induction of T helper cell type 1 and 17 responses. In conclusion, MPLA-tDCs are able to modulate antigen-specific responses of both naive and memory CD4+ T cells and might be a promising strategy to “turn off” self-reactive CD4+ effector T cells in autoimmunity.
Collapse
Affiliation(s)
- Jaxaira Maggi
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| | - Katina Schinnerling
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| | - Bárbara Pesce
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile , Santiago , Chile
| | - Catharien M Hilkens
- Musculoskeletal Research Group, Faculty of Medical Sciences, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Diego Catalán
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| | - Juan C Aguillón
- Programa Disciplinario de Inmunología, Immune Regulation and Tolerance Research Group, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy (MIII), Santiago, Chile
| |
Collapse
|
8
|
Krishnan B, Massilamany C, Basavalingappa RH, Rajasekaran RA, Kuszynski C, Switzer B, Peterson DA, Reddy J. Versatility of using major histocompatibility complex class II dextramers for derivation and characterization of antigen-specific, autoreactive T cell hybridomas. J Immunol Methods 2015; 426:86-94. [PMID: 26268454 PMCID: PMC4651793 DOI: 10.1016/j.jim.2015.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 01/09/2023]
Abstract
Antigen-specific, T cell hybridomas are useful to study the cellular, molecular and functional events, but their generation is a lengthy process. Thus, there is a need to develop robust methods to generate the hybridoma clones rapidly in a short period of time. To this end, we have demonstrated a novel approach using major histocompatibility complex (MHC) class II dextramers to generate T cell hybridomas for an autoantigen, proteolipid protein (PLP) 139-151. Using MHC class II dextramers assembled with PLP 139-151 as screening and sorting tools, we successfully obtained mono antigen-specific clones within seven to eight weeks. In conjunction with other T cell markers, dextramers permitted phenotypic characterization of hybridoma clones for their antigen specificity in a single step by flow cytometry. Importantly, we achieved successful fusions using dextramer(+) cells sorted by flow cytometry as a starting population, resulting in direct identification of multiple antigen-specific clones. Characterization of selected clones led us to identify chemokine receptor, CCR4(+) to be expressed consistently, but their cytokine-producing ability was variable. Our work provides a proof-of principle that the antigen-specific, CD4 T cell hybridoma clones can be generated directly using MHC class II dextramers. The availability of hybridoma clones that bind dextramers may serve as useful tools for various in vitro and in vivo applications.
Collapse
Affiliation(s)
- Bharathi Krishnan
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Chandirasegaran Massilamany
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Rakesh H Basavalingappa
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Rajkumar A Rajasekaran
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Charles Kuszynski
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Barbara Switzer
- College of Medicine, Dean's Research Laboratory, University of Nebraska Medical Center, Omaha, NE 68918, United States
| | - Daniel A Peterson
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
9
|
Deng L, Cho KJ, Fiers W, Saelens X. M2e-Based Universal Influenza A Vaccines. Vaccines (Basel) 2015; 3:105-36. [PMID: 26344949 PMCID: PMC4494237 DOI: 10.3390/vaccines3010105] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 12/23/2014] [Accepted: 01/30/2015] [Indexed: 12/13/2022] Open
Abstract
The successful isolation of a human influenza virus in 1933 was soon followed by the first attempts to develop an influenza vaccine. Nowadays, vaccination is still the most effective method to prevent human influenza disease. However, licensed influenza vaccines offer protection against antigenically matching viruses, and the composition of these vaccines needs to be updated nearly every year. Vaccines that target conserved epitopes of influenza viruses would in principle not require such updating and would probably have a considerable positive impact on global human health in case of a pandemic outbreak. The extracellular domain of Matrix 2 (M2e) protein is an evolutionarily conserved region in influenza A viruses and a promising epitope for designing a universal influenza vaccine. Here we review the seminal and recent studies that focused on M2e as a vaccine antigen. We address the mechanism of action and the clinical development of M2e-vaccines. Finally, we try to foresee how M2e-based vaccines could be implemented clinically in the future.
Collapse
Affiliation(s)
- Lei Deng
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Ki Joon Cho
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Walter Fiers
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| | - Xavier Saelens
- Inflammation Research Center, VIB, Technologiepark 927, B-9052 Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 927, B-9052 Ghent, Belgium.
| |
Collapse
|
10
|
Liu J, Wu B, Zhang S, Tan S, Sun Y, Chen Z, Qin Y, Sun M, Shi G, Wu Y, Sun M, Liu N, Ning K, Ma Y, Gao B, Yan J, Zhu F, Wang H, Gao GF. Conserved epitopes dominate cross-CD8+ T-cell responses against influenza A H1N1 virus among Asian populations. Eur J Immunol 2013; 43:2055-69. [PMID: 23681926 DOI: 10.1002/eji.201343417] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/05/2013] [Accepted: 05/13/2013] [Indexed: 12/12/2022]
Abstract
Novel strains of influenza A viruses (IAVs) have emerged with high infectivity and/or pathogenicity in recent years, causing worldwide concern. T cells are correlated with protection in humans through cross-reactive immunity against heterosubtypes of IAV. However, the different hierarchical roles of IAV-derived epitopes with distinct levels of polymorphism in the cross-reactive T-cell responses against IAV remain elusive. In this study, immunodominant epitopes scattered throughout the entire proteome of 2009 pandemic influenza A H1N1 virus and seasonal IAVs were synthesized and divided into different pools depending on their conservation. The overall profile of the IAV-specific CD8(+) T-cell immunity was detected by utilizing these peptide pools and also individual peptides. A dominant role of the conserved peptide-specific T-cell immunity was illuminated within the anti-IAV responses, while the CD8(+) T-cell responses against the variable epitopes were lower than the conserved peptides. As previously demonstrated within a Caucasian population, we determined that GL9-specific T cells, which also utilize Vβ 17 TCR (BV19), play a pivotal role in IAV-specific T-cell immunity within an HLA-A2(+) Asian population. Our study objectively reveals the different predominant roles of T-cell epitopes among IAV-specific cross-reactive cellular immunity. This may guide the development of vaccines with cross-T-cell immunogenicity against heterosubtypes of IAV.
Collapse
Affiliation(s)
- Jun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Bobadilla K, Sada E, Jaime ME, González Y, Ramachandra L, Rojas RE, Pedraza-Sánchez S, Michalak C, González-Noriega A, Torres M. Human phagosome processing of Mycobacterium tuberculosis antigens is modulated by interferon-γ and interleukin-10. Immunology 2013; 138:34-46. [PMID: 22924705 DOI: 10.1111/imm.12010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 07/24/2012] [Accepted: 08/22/2012] [Indexed: 01/14/2023] Open
Abstract
Intracellular pathogens, such as Mycobacterium tuberculosis, reside in the phagosomes of macrophages where antigenic processing is initiated. Mycobacterial antigen-MHC class II complexes are formed within the phagosome and are then trafficked to the cell surface. Interferon-γ (IFN-γ) and interleukin-10 (IL-10) influence the outcome of M. tuberculosis infection; however, the role of these cytokines with regard to the formation of M. tuberculosis peptide-MHC-II complexes remains unknown. We analysed the kinetics and subcellular localization of M. tuberculosis peptide-MHC-II complexes in M. tuberculosis-infected human monocyte-derived macrophages (MDMs) using autologous M. tuberculosis-specific CD4(+) T cells. The MDMs were pre-treated with either IFN-γ or IL-10 and infected with M. tuberculosis. Cells were mechanically homogenized, separated on Percoll density gradients and manually fractionated. The fractions were incubated with autologous M. tuberculosis -specific CD4(+) T cells. Our results demonstrated that in MDMs pre-treated with IFN-γ, M. tuberculosis peptide-MHC-II complexes were detected early mainly in the phagosomal fractions, whereas in the absence of IFN-γ, the complexes were detected in the endosomal fractions. In MDMs pre-treated with IL-10, the M. tuberculosis peptide-MHC-II complexes were retained in the endosomal fractions, and these complexes were not detected in the plasma membrane fractions. The results of immunofluorescence microscopy demonstrated the presence of Ag85B associated with HLA-DR at the cell surface only in the IFN-γ-treated MDMs, suggesting that IFN-γ may accelerate M. tuberculosis antigen processing and presentation at the cell membrane, whereas IL-10 favours the trafficking of Ag85B to vesicles that do not contain LAMP-1. Therefore, IFN-γ and IL-10 play a role in the formation and trafficking of M. tuberculosis peptide-MHC-II complexes.
Collapse
Affiliation(s)
- Karen Bobadilla
- Departamento de Microbiología, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
T cell hybridomas are very useful tools to investigate antigen presenting cell (APC) function. They were developed based on the fusion technology that led to monoclonal antibody section. Antigen-specific primary T cells are generated and fused to an immortal thymoma line. Unfused thymoma cells are eliminated by engineered metabolic selection. Antigen-specific hybridomas are identified and may be characterized in detail. Primary T cells are preferable for studies of the regulatory mechanisms intrinsic to T cells, but for study of antigen presentation T cell hybridomas have advantages over primary T cell clones, including their relative uniformity, stability over time, and ready availability in large numbers for extensive antigen presentation experiments.
Collapse
Affiliation(s)
- David H Canaday
- Division of Infectious Disease, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
13
|
HL C, R B, L J, TR H, PA H, CJ B, DH C. Characterization of MHC-II antigen presentation by B cells and monocytes from older individuals. Clin Immunol 2012; 144:172-7. [PMID: 22797466 PMCID: PMC3466105 DOI: 10.1016/j.clim.2012.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 05/22/2012] [Accepted: 06/11/2012] [Indexed: 01/27/2023]
Abstract
In this study we examine the effects of aging on antigen presentation of B cells and monocytes. We compared the antigen presentation function of peripheral blood B cells from young and old subjects using a system that specifically measures the B cell receptor (BCR)-mediated MHC-II antigen presentation. Monocytes were studied as well. Overall the mean magnitude of antigen presentation of soluble antigen and peptide was not different in older and younger subjects for both B cells and monocytes. Older subjects, however, showed increased heterogeneity of BCR-mediated antigen presentation by their B cells. The magnitude and variability of peptide presentation, which do not require uptake and processing, were the same between groups. Presentation by monocytes had similar variability between the older and younger subjects. These data suggest that poor B cell antigen processing, which results in diminished presentation in some older individuals may contribute to poor vaccine responses.
Collapse
Affiliation(s)
- Clark HL
- Division of Infectious Disease, Case Western Reserve University School of Medicine. 10900 Euclid Ave, BRB 1001, Cleveland, OH 44106-4984
| | - Banks R
- GRECC, Louis Stokes Cleveland VA Hospital, United States. 10701 East Boulevard Cleveland, OH 44106
| | - Jones L
- Division of Infectious Disease, Case Western Reserve University School of Medicine. 10900 Euclid Ave, BRB 1001, Cleveland, OH 44106-4984
- GRECC, Louis Stokes Cleveland VA Hospital, United States. 10701 East Boulevard Cleveland, OH 44106
| | - Hornick TR
- GRECC, Louis Stokes Cleveland VA Hospital, United States. 10701 East Boulevard Cleveland, OH 44106
| | - Higgins PA
- GRECC, Louis Stokes Cleveland VA Hospital, United States. 10701 East Boulevard Cleveland, OH 44106
- School of Nursing, Case Western Reserve University. 2120 Cornell Road Cleveland, Ohio 44106-4904
| | - Burant CJ
- GRECC, Louis Stokes Cleveland VA Hospital, United States. 10701 East Boulevard Cleveland, OH 44106
- School of Nursing, Case Western Reserve University. 2120 Cornell Road Cleveland, Ohio 44106-4904
| | - Canaday DH
- Division of Infectious Disease, Case Western Reserve University School of Medicine. 10900 Euclid Ave, BRB 1001, Cleveland, OH 44106-4984
- GRECC, Louis Stokes Cleveland VA Hospital, United States. 10701 East Boulevard Cleveland, OH 44106
| |
Collapse
|
14
|
Simmons DP, Wearsch PA, Canaday DH, Meyerson HJ, Liu YC, Wang Y, Boom WH, Harding CV. Type I IFN drives a distinctive dendritic cell maturation phenotype that allows continued class II MHC synthesis and antigen processing. THE JOURNAL OF IMMUNOLOGY 2012; 188:3116-26. [PMID: 22371391 DOI: 10.4049/jimmunol.1101313] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Microbial molecules or cytokines can stimulate dendritic cell (DC) maturation, which involves DC migration to lymph nodes and enhanced presentation of Ag to launch T cell responses. Microbial TLR agonists are the most studied inducers of DC maturation, but type I IFN (IFN-I) also promotes DC maturation. In response to TLR stimulation, DC maturation involves a burst of Ag processing with enhanced expression of peptide-class II MHC complexes and costimulator molecules. Subsequently, class II MHC (MHC-II) synthesis and expression in intracellular vacuolar compartments is inhibited, decreasing Ag processing function. This limits presentation to a cohort of Ags kinetically associated with the maturation stimulus and excludes presentation of Ags subsequently experienced by the DC. In contrast, our studies show that IFN-I enhances DC expression of MHC-II and costimulatory molecules without a concomitant inhibition of subsequent MHC-II synthesis and Ag processing. Expression of mRNA for MHC-II and the transcription factor CIITA is inhibited in DCs treated with TLR agonists but maintained in cells treated with IFN-I. After stimulation with IFN-I, MHC-II expression is increased on the plasma membrane but is also maintained in intracellular vacuolar compartments, consistent with sustained Ag processing function. These findings suggest that IFN-I drives a distinctive DC maturation program that enhances Ag presentation to T cells without a shutdown of Ag processing, allowing continued sampling of Ags for presentation.
Collapse
Affiliation(s)
- Daimon P Simmons
- Department of Pathology, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Bartley MB, Canaday DH. T cell hybridomas to study MHC-II restricted B-cell receptor-mediated antigen presentation by human B cells. J Immunol Methods 2011; 370:35-42. [PMID: 21620852 PMCID: PMC3308016 DOI: 10.1016/j.jim.2011.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 05/06/2011] [Accepted: 05/12/2011] [Indexed: 10/18/2022]
Abstract
MHC-II antigen presentation by B cells is essential in order for B cells to receive optimal costimulation from helper CD4+ T cells. This process is facilitated and focused through the extremely efficient uptake, processing, and presentation of antigen recognized by an individual B cell's unique B-cell receptor (BCR). The investigation of human B-cell antigen presentation has been limited by the varied specificity of BCR found in the mixed populations of B cells in vivo. As a result, there is no readily available method to measure BCR-mediated antigen presentation in this heterogeneous population of B cells. We have overcome this limitation by developing HLA-DR-restricted T-cell lines capable of recognizing a specific antigen taken up via the BCR and presented by the mixed B-cell population through this physiologically relevant mechanism. BCR-mediated presentation was enhanced >4 logs compared to presentation by B cells taking up the antigen through nonspecific mechanisms. The studies presented here characterize T-cell hybridoma lines developed for HLA-DRB1*0101+ and HLA-DRB1*1501+ B cells, but clones could be generated for other HLA-DR types using the methods described. These hybridomas have potential applications including study of the mechanisms of BCR-mediated enhancement of presentation, determination of adjuvant effects on presentation, and optimization of vaccine antigen preparations. Therefore, these T-cell lines could significantly facilitate the study of BCR-mediated antigen presentation required by T helper cell-dependent vaccines in humans.
Collapse
Affiliation(s)
- Matthew B. Bartley
- Division of Infectious Disease, Case Western Reserve University School of Medicine
| | - David H. Canaday
- Division of Infectious Disease, Case Western Reserve University School of Medicine
- Geriatric Research, Education and Clinical Center (GRECC), Cleveland VA
| |
Collapse
|
16
|
Ge X, James EA, Reijonen H, Kwok WW. Differences in self-peptide binding between T1D-related susceptible and protective DR4 subtypes. J Autoimmun 2011; 36:155-60. [PMID: 21257290 PMCID: PMC3046295 DOI: 10.1016/j.jaut.2010.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 12/17/2010] [Accepted: 12/22/2010] [Indexed: 10/18/2022]
Abstract
HLA-DR0401, 0403 and 0405 are associated with variable T1D susceptibilities when linked with a common HLA-DQ8 (DQA1∗0301/DQB1∗0302). It is unknown how the modest differences within the peptide binding regions of DR4 subtypes lead to distinct autoimmune risks. Since all Class II HLA molecules share the same intracellular compartments during biosynthesis, it is possible that DQ and DR compete with one another to bind and present antigenic peptides. As such, it is reasonable to hypothesize that a strong DR4 self-peptide binder down-modulates DQ8 epitope presentation more than a weak one. In this study, we first examined the binding of the peptides derived from two putative beta-cell autoantigens - GAD65 and insulin. Protective DR0403 bound similar number of self-peptides as susceptible DR0401 while highly susceptible DR0405 bound substantially less self-peptides than rest two molecules. Kinetic assays were used to further compare the stability of peptide:DR complexes formed between DR0401, 0403 and selected GAD65 peptides, which also bound DQ8. Two peptides with naturally processed DQ8 epitopes bound protective DR0403 with longer half-life and lower dissociation rate than susceptible DR0401, confirming DR0403 as a better peptide competitor than DR0401. The distinguishing peptide binding features of DR0401, DR0403, and DR0405 highlighted in this study help to explain the hierarchy of genetic associations between T1D and these DR4 subtypes. The enhanced peptide competition of DR0403 leads to a down-modulation of DQ8 epitope presentation, as compared to weak competitors such as DR0401 and DR0405, and therefore contributes to disease protection.
Collapse
Affiliation(s)
- Xinhui Ge
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
- Department of Immunology, University of Washington, Seattle, WA, 98195, USA
| | - Eddie A. James
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
| | - Helena Reijonen
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
- Department of Immunology, University of Washington, Seattle, WA, 98195, USA
| | - William W. Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
- Department of Immunology, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
17
|
Canaday DH, Burant CJ, Jones L, Aung H, Woc-Colburn L, Anthony DD. Preserved MHC-II antigen processing and presentation function in chronic HCV infection. Cell Immunol 2010; 266:187-91. [PMID: 21055734 PMCID: PMC3005840 DOI: 10.1016/j.cellimm.2010.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 10/04/2010] [Accepted: 10/12/2010] [Indexed: 01/07/2023]
Abstract
Individuals with chronic HCV infection have impaired response to vaccine, though the etiology remains to be elucidated. Dendritic cells (DC) and monocytes (MN) provide antigen uptake, processing, presentation, and costimulatory functions necessary to achieve optimal immune responses. The integrity of antigen processing and presentation function within these antigen presenting cells (APC) in the setting of HCV infection has been unclear. We used a novel T cell hybridoma system that specifically measures MHC-II antigen processing and presentation function of human APC. Results demonstrate MHC-II antigen processing and presentation function is preserved in both myeloid DC (mDC) and MN in the peripheral blood of chronically HCV-infected individuals, and indicates that an alteration in this function does not likely underlie the defective HCV-infected host response to vaccination.
Collapse
Affiliation(s)
- D H Canaday
- GRECC, Louis Stokes Cleveland VA Hospital, United States.
| | | | | | | | | | | |
Collapse
|
18
|
Mycobacterium tuberculosis promotes HIV trans-infection and suppresses major histocompatibility complex class II antigen processing by dendritic cells. J Virol 2010; 84:8549-60. [PMID: 20592078 DOI: 10.1128/jvi.02303-09] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mycobacterium tuberculosis is a leading killer of HIV-infected individuals worldwide, particularly in sub-Saharan Africa, where it is responsible for up to 50% of HIV-related deaths. Infection by HIV predisposes individuals to M. tuberculosis infection, and coinfection accelerates the progression of both diseases. In contrast to most other opportunistic infections associated with HIV, an increased risk of M. tuberculosis infection occurs during early-stage HIV disease, long before CD4 T cell counts fall below critical levels. We hypothesized that M. tuberculosis infection contributes to HIV pathogenesis by interfering with dendritic cell (DC)-mediated immune control. DCs carry pathogens like M. tuberculosis and HIV from sites of infection into lymphoid tissues, where they process and present antigenic peptides to CD4 T cells. Paradoxically, DCs can also deliver infectious HIV to T cells without first becoming infected, a process known as trans-infection. Lipopolysaccharide (LPS)-activated DCs sequester HIV in pocketlike membrane invaginations that remain open to the cell surface, and individual virions are delivered from the pocket into T cells at the site of contact during trans-infection. Here we report that M. tuberculosis exposure increases HIV trans-infection and induces viral sequestration within surface-accessible compartments identical to those seen in LPS-stimulated DCs. At the same time, M. tuberculosis dramatically decreases the degradative processing and major histocompatibility complex class II (MHC-II) presentation of HIV antigens to CD4 T cells. Our data suggest that M. tuberculosis infection promotes a shift in the dynamic balance between antigen processing and intact virion presentation, favoring DC-mediated amplification of HIV infections.
Collapse
|
19
|
Preserved MHC class II antigen processing in monocytes from HIV-infected individuals. PLoS One 2010; 5:e9491. [PMID: 20209134 PMCID: PMC2831061 DOI: 10.1371/journal.pone.0009491] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 12/21/2009] [Indexed: 11/24/2022] Open
Abstract
Background MHC-II restricted CD4+ T cells are dependent on antigen presenting cells (APC) for their activation. APC dysfunction in HIV-infected individuals could accelerate or exacerbate CD4+ T cell dysfunction and may contribute to increased levels of immunodeficiency seen in some patients regardless of their CD4+ T cell numbers. Here we test the hypothesis that APC from HIV-infected individuals have diminished antigen processing and presentation capacity. Methodology/Principal Findings Monocytes (MN) were purified by immuno-magnetic bead isolation techniques from HLA-DR1.01+ or DR15.01+ HIV-infected and uninfected individuals. MN were analyzed for surface MHC-II expression and for antigen processing and presentation capacity after overnight incubation with soluble antigen or peptide and HLA-DR matched T cell hybridomas. Surface expression of HLA-DR was 20% reduced (p<0.03) on MN from HIV-infected individuals. In spite of this, there was no significant difference in antigen processing and presentation by MN from 14 HIV-infected donors (8 HLA-DR1.01+ and 6 HLA-DR15.01+) compared to 24 HIV-uninfected HLA-matched subjects. Conclusions/Significance We demonstrated that MHC class II antigen processing and presentation is preserved in MN from HIV-infected individuals. This further supports the concept that this aspect of APC function does not further contribute to CD4+ T cell dysfunction in HIV disease.
Collapse
|
20
|
Harding CV, Canaday D, Ramachandra L. Choosing and preparing antigen-presenting cells. CURRENT PROTOCOLS IN IMMUNOLOGY 2010; Chapter 16:16.1.1-16.1.30. [PMID: 20143315 DOI: 10.1002/0471142735.im1601s88] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The first issue in selecting a system for antigen-presentation experiments is to define the appropriate type of antigen-presenting cell (APC) to study. For some experiments, crude preparations such as splenocytes or peripheral blood mononuclear cells (PBMCs) may suffice to provide APC function for stimulating T cells. This unit develops approaches for preparation of more defined APC populations, including dendritic cells (DCs), macrophages, and B lymphocytes, the three types of "professional" APC. Each of these cell types exists in different stages of differentiation, maturation, and activation, or in some cases different lineages. For example, dendritic cells may be divided into subsets, including myeloid DCs (mDCs) and plasmacytoid DCs (pDCs). Each APC type has an important antigen-presentation function, although they contribute to different aspects of the immune response. Therefore, selection of an APC type for study must include consideration of the stage or aspect of immune response that is to be modeled in the experiment.
Collapse
|
21
|
Harding CV, Ramachandra L. Presenting exogenous antigen to T cells. CURRENT PROTOCOLS IN IMMUNOLOGY 2010; Chapter 16:16.2.1-16.2.18. [PMID: 20143316 DOI: 10.1002/0471142735.im1602s88] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Antigen processing and presentation experiments can be done with a wide variety of antigen-presenting cells (APCs). Most experiments will use one of the "professional" APC types: dendritic cells (DCs), macrophages, and B lymphocytes. Other types of cells may be used for antigen presentation in some circumstances. Each type of professional APC has an important antigen-presentation function, but the different APC types contribute to different aspects of the immune response. Therefore, selection of an APC type for study must include consideration of the stage or aspect of immune response that is to be modeled in the experiment. An important technical distinction for some types of experiments is whether the APCs are adherent or nonadherent, since this dictates the procedures that must be used to wash the cells as the medium is changed.
Collapse
|
22
|
PLGA microparticles in respirable sizes enhance an in vitro T cell response to recombinant Mycobacterium tuberculosis antigen TB10.4-Ag85B. Pharm Res 2009; 27:350-60. [PMID: 20024670 DOI: 10.1007/s11095-009-0028-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Accepted: 12/03/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE To study the use of poly (lactide-co-glycolide) (PLGA) microparticles in respirable sizes as carriers for recombinant tuberculosis (TB) antigen, TB10.4-Ag85B, with the ultimate goal of pulmonary delivery as vaccine for the prevention of TB. MATERIALS AND METHODS Recombinant TB antigens were purified from E. coli by FPLC and encapsulated into PLGA microparticles by emulsion/spray-drying. Spray-drying condition was optimized by half-factorial design. Microparticles encapsulating TB antigens were assessed for their ability to deliver antigens to macrophages for subsequent presentation by employing an in vitro antigen presentation assay specific to an Ag85B epitope. RESULTS Spray-drying condition was optimized to prepare PLGA microparticles suitable for pulmonary delivery (aerodynamic diameter of 3.3 microm). Antigen release from particles exhibited an initial burst release followed by sustained release up to 10 days. Antigens encapsulated into PLGA microparticles induced much stronger interleukin-2 secretion in a T-lymphocyte assay compared to antigen solutions for three particle formulations. Macrophages pulsed with PLGA-MDP-TB10.4-Ag85B demonstrated extended epitope presentation. CONCLUSION PLGA microparticles in respirable sizes were effective in delivering recombinant TB10.4-Ag85B in an immunologically relevant manner to macrophages. These results set the foundation for further investigation into the potential use of PLGA particles for pulmonary delivery of vaccines to prevent Mycobacterium tuberculosis infection.
Collapse
|
23
|
Wu Y, Wu W, Wong WM, Ward E, Thrasher AJ, Goldblatt D, Osman M, Digard P, Canaday DH, Gustafsson K. Human gamma delta T cells: a lymphoid lineage cell capable of professional phagocytosis. THE JOURNAL OF IMMUNOLOGY 2009; 183:5622-9. [PMID: 19843947 DOI: 10.4049/jimmunol.0901772] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Professional phagocytosis in mammals is considered to be performed exclusively by myeloid cell types. In this study, we demonstrate, for the first time, that a mammalian lymphocyte subset can operate as a professional phagocyte. By using confocal microscopy, transmission electron microscopy, and functional Ag presentation assays, we find that freshly isolated human peripheral blood gammadelta T cells can phagocytose Escherichia coli and 1 microm synthetic beads via Ab opsonization and CD16 (FcgammaRIII), leading to Ag processing and presentation on MHC class II. In contrast, other CD16(+) lymphocytes, i.e., CD16(+)/CD56(+) NK cells, were not capable of such functions. These findings of distinct myeloid characteristics in gammadelta T cells strongly support the suggestion that gammadelta T cells are evolutionarily ancient lymphocytes and have implications for our understanding of their role in transitional immunity and the control of infectious diseases and cancer.
Collapse
Affiliation(s)
- Yin Wu
- Molecular Immunology Unit, University College London Institute of Child Health, London, U.K
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Astiazaran-Garcia H, Quintero J, Vega R, Briceño P, Oviedo C, Rascon L, Garibay-Escobar A, Castillo-Yañez FJ, Robles-Zepeda R, Hernandez J, Velazquez C. Identification of T-cell stimulating antigens from Giardia lamblia by using Giardia-specific T-cell hybridomas. Parasite Immunol 2009; 31:132-9. [PMID: 19222784 DOI: 10.1111/j.1365-3024.2008.01083.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
T-cell immune response plays an important role in controlling Giardia lamblia infections. Little is known about the G. lamblia-specific antigens that stimulate a cell-mediated immune response. The aim of the present study was to identify T-cell stimulating G. lamblia antigens. For this purpose, we generated a group of Giardia-specific T-cell hybridomas (2F9, 4D5, 6D10, 8B9, 9B10, 10F7 and 10G5). Hybridomas were screened for reactivity with G. lamblia protein extract by the CTLL bioassay. These T-cell hybridomas did not exhibit any significant activation either in the absence of G. lamblia protein extract or in the presence of irrelevant antigen (hen white egg lysozyme). To further characterize the T-cell hybridomas generated, we selected three hybridomas (10G5, 4D5 and 9B10). Giardia lamblia proteins of 90-110, 65-77 and 40-64 kDa showed T-cell stimulating activity for the hybridomas 10G5, 4D5 and 9B10, respectively, in a concentration-dependent manner. Protein extract obtained from different G. lamblia strains (GS/M-83-H7, WB C6 and a clinical isolate (YJJ)) stimulated all T-cell hybridomas, indicating that T-cell-stimulating antigens are expressed among different G. lamblia strains. In conclusion, we identified T-cell stimulating G. lamblia antigens by using Giardia-specific T-cell hybridomas. To our knowledge, these hybridomas are the first-described T-cell hybridomas specific for G. lamblia.
Collapse
Affiliation(s)
- H Astiazaran-Garcia
- Centro de Investigación en Alimentación y Desarrollo A.C. Hermosillo, Sonora, México
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Endothelial Progenitor Cells Possess Monocyte-like Antigen-presenting and T-cell-Co-stimulatory Capacity. Transplantation 2009; 87:340-9. [DOI: 10.1097/tp.0b013e3181957308] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
26
|
Immunodominance in mouse and human CD4+ T-cell responses specific for the Bordetella pertussis virulence factor P.69 pertactin. Infect Immun 2008; 77:896-903. [PMID: 19015250 DOI: 10.1128/iai.00769-08] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
P.69 pertactin (P.69 Prn), an adhesion molecule from the causative agent of pertussis, Bordetella pertussis, is present in cellular and most acellular vaccines that are currently used worldwide. Although both humoral immunity and cellular immunity directed against P.69 Prn have been implicated in protective immune mechanisms, the identities of CD4(+) T-cell epitopes on the P.69 Prn protein remain unknown. Here, a single I-A(d)-restricted B. pertussis conserved CD4(+) T-cell epitope at the N terminus of P.69 Prn was identified by using a BALB/c T-cell hybridoma. The epitope appeared immunodominant among four other minor strain-conserved P.69 Prn epitopes recognized after vaccination and B. pertussis infection, and it was capable of evoking a Th1/Th17-type cytokine response. B. pertussis P.69 Prn immune splenocytes did not cross-react with natural variants of the epitope as present in Bordetella parapertussis and Bordetella bronchiseptica. Finally, it was found that the immunodominant P.69 Prn epitope is broadly recognized in the human population by CD4(+) T cells in an HLA-DQ-restricted manner. During B. pertussis infection, the epitope was associated with a Th1-type CD4(+) T-cell response. Hence, this novel P.69 Prn epitope is involved in CD4(+) T-cell immunity after B. pertussis vaccination and infection in mice and, more importantly, in humans. Thus, it may provide a useful tool for the evaluation of the type, magnitude, and maintenance of B. pertussis-specific CD4(+) T-cell mechanisms in preclinical and clinical vaccine studies.
Collapse
|
27
|
Vasilevsky S, Colino J, Puliaev R, Canaday DH, Snapper CM. Macrophages pulsed with Streptococcus pneumoniae elicit a T cell-dependent antibody response upon transfer into naive mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:1787-97. [PMID: 18641316 PMCID: PMC2561269 DOI: 10.4049/jimmunol.181.3.1787] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Macrophages are less effective than DC at priming naive CD4(+) T cells, suggesting that DC are unique in initiating T cell-dependent Ab responses. We compared the ability of DC and macrophages, pulsed in vitro with Streptococcus pneumoniae, to elicit protein- and polysaccharide-specific Ig isotype production upon adoptive transfer into naive mice. S. pneumoniae-activated DC secreted more proinflammatory and anti-inflammatory cytokines, expressed higher levels of surface MHC class II and CD40, and presented S. pneumoniae or recombinant pneumococcal surface protein A (PspA) to a PspA-specific T hybridoma more efficiently than macrophages. However, upon adoptive transfer into naive mice, S. pneumoniae-pulsed macrophages elicited an IgM or IgG anti-PspA and anti-polysaccharide response comparable in serum titers and IgG isotype distribution to that induced by DC. The IgG anti-PspA response, in contrast to the IgG anti-polysaccharide, to S. pneumoniae-pulsed macrophages was T cell-dependent. S. pneumoniae-pulsed macrophages that were paraformaldehyde-fixed before transfer or lacking expression of MHC class II or CD40 were highly defective in eliciting an anti-PspA response, although the anti-polysaccharide response was largely unaffected. To our knowledge, these data are the first to indicate that macrophages can play an active role in the induction of a T cell-dependent humoral immune response in a naive host.
Collapse
Affiliation(s)
- Sam Vasilevsky
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Jesus Colino
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Roman Puliaev
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - David H. Canaday
- Division of Infectious Disease, Case Western Reserve University, Cleveland, OH 44106
| | - Clifford M. Snapper
- Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| |
Collapse
|
28
|
Soualhine H, Deghmane AE, Sun J, Mak K, Talal A, Av-Gay Y, Hmama Z. Mycobacterium bovis bacillus Calmette-Guérin secreting active cathepsin S stimulates expression of mature MHC class II molecules and antigen presentation in human macrophages. THE JOURNAL OF IMMUNOLOGY 2007; 179:5137-45. [PMID: 17911599 DOI: 10.4049/jimmunol.179.8.5137] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A successful Th cell response to bacterial infections is induced by mature MHC class II molecules presenting specific Ag peptides on the surface of macrophages. In recent studies, we demonstrated that infection with the conventional vaccine Mycobacterium bovis bacillus Calmette-Guérin (BCG) specifically blocks the surface export of mature class II molecules in human macrophages by a mechanism dependent on inhibition of cathepsin S (Cat S) expression. The present study examined class II expression in macrophages infected with a rBCG strain engineered to express and secrete biologically active human Cat S (rBCG-hcs). Cat S activity was completely restored in cells ingesting rBCG-hcs, which secreted substantial levels of Cat S intracellularly. Thus, infection with rBCG-hcs, but not parental BCG, restored surface expression of mature MHC class II molecules in response to IFN-gamma, presumably as result of MHC class II invariant chain degradation dependent on active Cat S secreted by the bacterium. These events correlated with increased class II-directed presentation of mycobacterial Ag85B to a specific CD4(+) T cell hybridoma by rBCG-hcs-infected macrophages. Consistent with these findings, rBCG-hcs was found to accelerate the fusion of its phagosome with lysosomes, a process that optimizes Ag processing in infected macrophages. These data demonstrated that intracellular restoration of Cat S activity improves the capacity of BCG-infected macrophages to stimulate CD4(+) Th cells. Given that Th cells play a major role in protection against tuberculosis, rBCG-hcs would be a valuable tuberculosis vaccine candidate.
Collapse
Affiliation(s)
- Hafid Soualhine
- Division of Infectious Diseases, Department of Medicine, University of British Columbia and Vancouver Costal Health Institute, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | |
Collapse
|
29
|
Lu D, Garcia-Contreras L, Xu D, Kurtz SL, Liu J, Braunstein M, McMurray DN, Hickey AJ. Poly (lactide-co-glycolide) microspheres in respirable sizes enhance an in vitro T cell response to recombinant Mycobacterium tuberculosis antigen 85B. Pharm Res 2007; 24:1834-43. [PMID: 17657598 DOI: 10.1007/s11095-007-9302-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2007] [Accepted: 03/20/2007] [Indexed: 11/29/2022]
Abstract
PURPOSE To investigate the use of poly (lactide-co-glycolide) (PLGA) microparticles in respirable sizes as carriers for Antigen 85B (Ag85B), a secreted protein of Mycobacterium tuberculosis, with the ultimate goal of employing them in pulmonary delivery of tuberculosis vaccine. MATERIALS AND METHODS Recombinant Ag85B was expressed from two Escherichia coli strains and encapsulated by spray-drying in PLGA microspheres with/without adjuvants. These microspheres containing rAg85B were assessed for their ability to deliver antigen to macrophages for subsequent processing and presentation to the specific CD4 T-hybridoma cells DB-1. DB-1 cells recognize the Ag85B(97-112) epitope presented in the context of MHC class II and secrete IL-2 as the cytokine marker. RESULTS Microspheres suitable for aerosol delivery to the lungs (3.4-4.3 microm median diameter) and targeting alveolar macrophages were manufactured. THP-1 macrophage-like cells exposed with PLGA-rAg85B microspheres induced the DB-1 cells to produce IL-2 at a level that was two orders of magnitude larger than the response elicited by soluble rAg85B. This formulation demonstrated extended epitope presentation. CONCLUSIONS PLGA microspheres in respirable sizes were effective in delivering rAg85B in an immunologically relevant manner to macrophages. These results are a foundation for further investigation into the potential use of PLGA particles for delivery of vaccines to prevent M. tuberculosis infection.
Collapse
Affiliation(s)
- Dongmei Lu
- Molecular Pharmaceutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7360, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Jones L, McDonald D, Canaday DH. Rapid MHC-II antigen presentation of HIV type 1 by human dendritic cells. AIDS Res Hum Retroviruses 2007; 23:812-6. [PMID: 17604545 DOI: 10.1089/aid.2006.0280] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
HIV-1 is taken up by dendritic cells (DC) and degradation is detectable within a few hours. Little is known about how rapidly HIV-peptide-loaded MHC-II complexes appear on the surface of DC, however. A key impediment to the detailed understanding of MHC-II antigen presentation of HIV-1 has been the lack of good tools to quantitatively measure antigen presentation. We have developed HIV-1-gag p24 and reverse transcriptase (RT)-specific CD4(+) T cell hybridomas that demonstrate high sensitivity and specificity to detect HIV-1 antigens presented by MHC-II on human DC. We demonstrate that ex vivo primary blood myeloid DC (mDC) and monocyte-derived DC (MDDC) presented HIV-1 peptides on MHC-II molecules within 2 h of exposure to virions. HIV-1 was degraded in a compartment requiring acidification for processing and then was loaded onto newly synthesized MHC-II molecules for presentation.
Collapse
Affiliation(s)
- Leola Jones
- Division of Infectious Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-4984, USA
| | | | | |
Collapse
|
31
|
Richards KA, Chaves FA, Krafcik FR, Topham DJ, Lazarski CA, Sant AJ. Direct ex vivo analyses of HLA-DR1 transgenic mice reveal an exceptionally broad pattern of immunodominance in the primary HLA-DR1-restricted CD4 T-cell response to influenza virus hemagglutinin. J Virol 2007; 81:7608-19. [PMID: 17507491 PMCID: PMC1933370 DOI: 10.1128/jvi.02834-06] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The recent threat of an avian influenza pandemic has generated significant interest in enhancing our understanding of the events that dictate protective immunity to influenza and in generating vaccines that can induce heterosubtypic immunity. Although antigen-specific CD4 T cells are known to play a key role in protective immunity to influenza through the provision of help to B cells and CD8 T cells, little is known about the specificity and diversity of CD4 T cells elicited after infection, particularly those elicited in humans. In this study, we used HLA-DR transgenic mice to directly and comprehensively identify the specificities of hemagglutinin (HA)-specific CD4 T cells restricted to a human class II molecule that were elicited following intranasal infection with a strain of influenza virus that has been endemic in U.S. human populations for the last decade. Our results reveal a surprising degree of diversity among influenza virus-specific CD4 T cells. As many as 30 different peptides, spanning the entire HA protein, were recognized by CD4 T cells, including epitopes genetically conserved among H1, H2, and H5 influenza A viruses. We also compared three widely used major histocompatibility class II algorithms to predict HLA-DR binding peptides and found these as yet inadequate for identifying influenza virus-derived epitopes. The results of these studies offer key insights into the spectrum of peptides recognized by HLA-DR-restricted CD4 T cells that may be the focus of immune responses to infection or to experimental or clinical vaccines in humans.
Collapse
Affiliation(s)
- Katherine A Richards
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
32
|
Canaday DH, Chakravarti S, Srivastava T, Tisch DJ, Cheruvu VK, Harding CV, Ramachandra L. Class II MHC antigen presentation defect in neonatal monocytes is not correlated with decreased MHC-II expression. Cell Immunol 2006; 243:96-106. [PMID: 17324388 PMCID: PMC1904503 DOI: 10.1016/j.cellimm.2007.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2006] [Revised: 01/05/2007] [Accepted: 01/08/2007] [Indexed: 10/23/2022]
Abstract
Neonates are at increased risk of infections compared to adults. To dissect the mechanisms that contribute to neonatal immune deficiency, we compared MHC-II antigen processing and presentation by monocytes from umbilical cord blood and unrelated adult controls. Antigen-specific, co-stimulation-independent murine T hybridoma cells were used to detect peptide:HLA-DR complexes. Relative to adult monocytes, neonatal monocytes were significantly defective in processing and presentation of protein antigens and presentation of exogenous peptide. Defects in responses to protein antigens and exogenous peptide were of similar magnitude (56-81% decrease), indicating that the defect lies in antigen presentation as opposed to intracellular antigen processing. Average surface MHC-II levels on neonatal monocytes were 38% less than on adult monocytes. However, there was no correlation between decreased MHC-II expression on individual neonatal monocyte samples and reduced T cell responses. We demonstrate for the first time that neonatal monocytes are defective in MHC-II antigen presentation by a mechanism not correlated with decreased MHC-II expression.
Collapse
Affiliation(s)
- David H. Canaday
- Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Soma Chakravarti
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH
- Rainbow Babies and Children’s Hospital, Cleveland, OH
| | - Tarun Srivastava
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Daniel J. Tisch
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH
| | - Vinay K. Cheruvu
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH
| | | | - Lakshmi Ramachandra
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH
- Department of Pathology, Case Western Reserve University, Cleveland, OH
- Rainbow Babies and Children’s Hospital, Cleveland, OH
| |
Collapse
|
33
|
Pang KC, Wei JQZ, Chen W. Dynamic quantification of MHC class I–peptide presentation to CD8+ T cells via intracellular cytokine staining. J Immunol Methods 2006; 311:12-8. [PMID: 16516224 DOI: 10.1016/j.jim.2006.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Revised: 12/22/2005] [Accepted: 01/03/2006] [Indexed: 11/25/2022]
Abstract
In order to further our basic understanding of antigen processing and presentation as well as to translate that knowledge into clinically effective vaccines and immunotherapies, having appropriate tools to study MHC class I-peptide presentation is highly desirable. Current methods are based upon HPLC fractionation of extracted peptides, monoclonal Ab, multivalent T cell receptors (TCR), T cell hybridomas, TCR transgenic cells, and T cell lines. However, each of these is associated with problems that make them either difficult to apply generally or too insensitive to adequately quantitate antigen presentation. We have developed a method based upon intracellular cytokine staining (ICS) that dynamically and relatively quantitates MHC class I-peptide presentation to CD8+ T cells in a manner that is both widely applicable and highly sensitive. It is well-suited to assess antigen presentation in its early stages, does not require fixation nor labeling of antigen presenting cells (APC), can be used to examine cross-presentation, and is able to directly employ ex vivo T cells which obviates the need for the development and maintenance of T cell lines and hybridomas. Our method represents a simple yet powerful tool that others interested in studying antigen processing and presentation should find of great practical value.
Collapse
Affiliation(s)
- Ken C Pang
- T cell Laboratory, Ludwig Institute for Cancer Research, Melbourne Branch, Austin Health, Heidelberg, VIC, 3084, Australia
| | | | | |
Collapse
|
34
|
Torres M, Ramachandra L, Rojas RE, Bobadilla K, Thomas J, Canaday DH, Harding CV, Boom WH. Role of phagosomes and major histocompatibility complex class II (MHC-II) compartment in MHC-II antigen processing of Mycobacterium tuberculosis in human macrophages. Infect Immun 2006; 74:1621-30. [PMID: 16495533 PMCID: PMC1418651 DOI: 10.1128/iai.74.3.1621-1630.2006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Revised: 10/13/2005] [Accepted: 12/07/2005] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium tuberculosis resides in phagosomes inside macrophages. In this study, we analyzed the kinetics and location of M. tuberculosis peptide-major histocompatibility complex class II (MHC-II) complexes in M. tuberculosis-infected human macrophages. M. tuberculosis peptide-MHC-II complexes were detected with polyclonal autologous M. tuberculosis-specific CD4+ T cells or F9A6 T hybridoma cells specific for M. tuberculosis antigen (Ag) 85B (96-111). Macrophages processed heat-killed M. tuberculosis more rapidly and efficiently than live M. tuberculosis. To determine where M. tuberculosis peptide-MHC-II complexes were formed intracellularly, macrophages incubated with heat-killed M. tuberculosis were homogenized, and subcellular compartments were separated on Percoll density gradients analyzed with T cells. In THP-1 cells, M. tuberculosis Ag 85B (96- 111)-DR1 complexes appeared initially in phagosomes, followed by MHC class II compartment (MIIC) and the plasma membrane fractions. In monocyte-derived macrophages, M. tuberculosis peptide-MHC-II complexes appeared only in MIIC fractions and subsequently on the plasma membrane. Although phagosomes from both cell types acquired lysosome-associated membrane protein 1 (LAMP-1) and MHC-II, THP-1 phagosomes that support formation of M. tuberculosis peptide-MHC-II complexes had increased levels of both LAMP-1 and MHC-II. Thus, M. tuberculosis phagosomes with high levels of MHC-II and LAMP-1 and MIIC both have the potential to form peptide-MHC-II complexes from M. tuberculosis antigens in human macrophages.
Collapse
Affiliation(s)
- Martha Torres
- Division of Infectious Diseases, Dept. of Pediatrics, Rainbow Babies and Children's Hospital, Rm. 4007, 2101 Adelbert Rd., Cleveland, OH 44106-6008, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Stagg J, Pommey S, Eliopoulos N, Galipeau J. Interferon-gamma-stimulated marrow stromal cells: a new type of nonhematopoietic antigen-presenting cell. Blood 2005; 107:2570-7. [PMID: 16293599 DOI: 10.1182/blood-2005-07-2793] [Citation(s) in RCA: 232] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Several studies have demonstrated that marrow stromal cells (MSCs) can suppress allogeneic T-cell responses. However, the effect of MSCs on syngeneic immune responses has been largely overlooked. We describe here that primary MSCs derived from C57BL/6 mice behave as conditional antigen-presenting cells (APCs) and can induce antigen-specific protective immunity. Interferon gamma (IFNgamma)-treated C57BL/6 MSCs, but not unstimulated MSCs, cocultured with ovalbumin-specific major histocompatibility (MHC) class II-restricted hybridomas in the presence of soluble ovalbumin-induced significant production of interleukin-2 (IL-2) in an antigen dose-dependent manner (P < .005). IFNgamma-treated MSCs could further activate in vitro ovalbumin-specific primary transgenic CD4+ T cells. C57BL/6 MSCs, however, were unable to induce antigen cross-presentation via the MHC class I pathway. When syngeneic mice were immunized intraperitoneally with ovalbumin-pulsed IFNgamma-treated MSCs, they developed antigen-specific cytotoxic CD8+ T cells and became fully protected (10 of 10 mice) against ovalbumin-expressing E.G7 tumors. Human MSCs were also studied for antigen-presenting functions. IFNgamma-treated DR1-positive human MSCs, but not unstimulated human MSCs, induced significant production of IL-2 when cocultured with DR1-restricted influenza-specific humanized T-cell hybridomas in the presence of purified influenza matrix protein 1. Taken together, our data strongly suggest that MSCs behave as conditional APCs in syngeneic immune responses.
Collapse
Affiliation(s)
- John Stagg
- Lady Davis Institute for Medical Research, 3755 Cote Ste-Catherine Rd, Montreal, QC, Canada H3T 1E2
| | | | | | | |
Collapse
|
36
|
Bagai R, Valujskikh A, Canaday DH, Bailey E, Lalli PN, Harding CV, Heeger PS. Mouse endothelial cells cross-present lymphocyte-derived antigen on class I MHC via a TAP1- and proteasome-dependent pathway. THE JOURNAL OF IMMUNOLOGY 2005; 174:7711-5. [PMID: 15944272 DOI: 10.4049/jimmunol.174.12.7711] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In vivo studies suggest that vascular endothelial cells (ECs) can acquire and cross-present exogenous Ag on MHC-I but the cellular mechanisms underlying this observation remain unknown. We tested whether primary female mouse aortic ECs could cross-present exogenous male Ag to the T cell hybridoma, MHH, specific for HYUty plus D(b). MHC-I-deficient male spleen cells provided a source of male Ag that could not directly stimulate the MHH cells. Addition of male but not female MHC-I-deficient spleen cells to wild-type syngeneic female EC induced MHH stimulation, demonstrating EC cross-presentation. Lactacystin treatment of the donor male MHC-I-deficient spleen cells, to inhibit proteasome function, markedly enhanced EC cross-presentation showing that the process is most efficient for intact proteins rather than degraded peptide fragments. Additional experiments revealed that this EC Ag-processing pathway is both proteasome and TAP1 dependent. These studies demonstrate that cultured murine aortic ECs can process and present MHC-I-restricted Ag derived from a separate, live cell, and they offer insight into the molecular requirements involved in this EC Ag presentation process. Through this pathway, ECs expressing cross-presented peptides can participate in the effector phase of T cell-mediated inflammatory responses such as autoimmunity, anti-tumor immunity, and transplant rejection.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2
- ATP-Binding Cassette Transporters/physiology
- Animals
- Antigen Presentation
- Cells, Cultured
- Cross-Priming/immunology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Female
- H-Y Antigen/genetics
- H-Y Antigen/immunology
- H-Y Antigen/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Hybridomas
- Intracellular Fluid/immunology
- Intracellular Fluid/metabolism
- Male
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Minor Histocompatibility Antigens
- Proteasome Endopeptidase Complex/physiology
- Proteins/genetics
- Proteins/immunology
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Rakesh Bagai
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Tobian AAR, Harding CV, Canaday DH. Mycobacterium tuberculosis heat shock fusion protein enhances class I MHC cross-processing and -presentation by B lymphocytes. THE JOURNAL OF IMMUNOLOGY 2005; 174:5209-14. [PMID: 15843516 DOI: 10.4049/jimmunol.174.9.5209] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Exogenous heat shock protein (HSP):peptide complexes are processed for cross-presentation of HSP-chaperoned peptides on class I MHC (MHC-I) molecules. Fusion proteins containing HSP and Ag sequences facilitate MHC-I cross-presentation of linked antigenic epitopes. Processing of HSP-associated Ag has been attributed to dendritic cells and macrophages. We now provide the first evidence to show processing of HSP-associated Ag for MHC-I cross-presentation by B lymphocytes. Fusion of OVA sequence (rOVA, containing OVA(230-359) sequence) to Mycobacterium tuberculosis HSP70 greatly enhanced rOVA processing and MHC-I cross-presentation of OVA(257-264):K(b) complexes by B cells. Enhanced processing was dependent on linkage of rOVA sequence to HSP70. M. tuberculosis HSP70-OVA fusion protein enhanced cross-processing by a CD91-dependent process that was independent of TLR4 and MyD88. The enhancement occurred through a post-Golgi, proteasome-independent mechanism. These results indicate that HSPs enhance delivery and cross-processing of HSP-linked Ag by B cells, which could provide a novel contribution to the generation of CD8(+) T cell responses. HSP fusion proteins have potential advantages for use in vaccines to enhance priming of CD8(+) T cell responses.
Collapse
Affiliation(s)
- Aaron A R Tobian
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
38
|
Zhou D, Li P, Lin Y, Lott JM, Hislop AD, Canaday DH, Brutkiewicz RR, Blum JS. Lamp-2a facilitates MHC class II presentation of cytoplasmic antigens. Immunity 2005; 22:571-81. [PMID: 15894275 DOI: 10.1016/j.immuni.2005.03.009] [Citation(s) in RCA: 227] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 02/28/2005] [Accepted: 03/16/2005] [Indexed: 11/28/2022]
Abstract
Extracellular antigens are internalized and processed before binding MHC class II molecules within endosomal and lysosomal compartments of professional antigen presenting cells (APC) for subsequent presentation to T cells. Yet select cytoplasmic peptides derived from autoantigens also intersect and bind class II molecules via an unknown mechanism. In human B lymphoblasts, inhibition of the peptide transporter associated with antigen processing (TAP) failed to alter class II-restricted cytoplasmic epitope presentation. By contrast, decreased display of cytoplasmic epitopes via class II molecules was observed in cells with diminished expression of the lysosome-associated membrane protein-2 (Lamp-2). Overexpression of Lamp-2 isoform A (Lamp-2a), an established component of chaperone-mediated autophagy, enhanced cytoplasmic autoantigen presentation. Manipulating APC expression of heat shock cognate protein 70 (hsc70), a cofactor for Lamp-2a, also altered cytoplasmic class II peptide presentation. These results demonstrate a novel role for the lysosomal Lamp-2a-hsc70 complex in promoting immunological recognition and antigen presentation.
Collapse
Affiliation(s)
- Delu Zhou
- Center for Immunobiology, Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Gehring AJ, Dobos KM, Belisle JT, Harding CV, Boom WH. Mycobacterium tuberculosis LprG (Rv1411c): a novel TLR-2 ligand that inhibits human macrophage class II MHC antigen processing. THE JOURNAL OF IMMUNOLOGY 2004; 173:2660-8. [PMID: 15294983 DOI: 10.4049/jimmunol.173.4.2660] [Citation(s) in RCA: 193] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
MHC class II (MHC-II)-restricted CD4(+) T cells are essential for control of Mycobacterium tuberculosis infection. This report describes the identification and purification of LprG (Rv1411c) as an inhibitor of primary human macrophage MHC-II Ag processing. LprG is a 24-kDa lipoprotein found in the M. tuberculosis cell wall. Prolonged exposure (>16 h) of human macrophages to LprG resulted in marked inhibition of MHC-II Ag processing. Inhibition of MHC-II Ag processing was dependent on TLR-2. Short-term exposure (<6 h) to LprG stimulated TLR-2-dependent TNF-alpha production. Thus, LprG can exploit TLR-2 signaling to inhibit MHC-II Ag processing in human macrophages. Inhibition of MHC-II Ag processing by mycobacterial lipoproteins may allow M. tuberculosis, within infected macrophages, to avoid recognition by CD4(+) T cells.
Collapse
Affiliation(s)
- Adam J Gehring
- Division of Infectious Diseases, Case Western Reserve University and University Hospitals of Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|