1
|
Sundebo Meldgaard T, Viborg N, Suarez Hernandez S, Vazquez Albacete D, Tamhane T, Reker Hadrup S. Validation of novel conditional ligands and large-scale detection of antigen-specific T cells for H-2D d and H-2K d. Sci Rep 2024; 14:12292. [PMID: 38811654 PMCID: PMC11136991 DOI: 10.1038/s41598-024-62938-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024] Open
Abstract
The UV-mediated peptide exchange has enabled the generation of multiple different MHC multimer specificities in parallel, surpassing tedious individual refolding of MHC molecules with peptide ligands. Murine models are acknowledged as an effective tool for preclinical research to advance our understanding of immunological mechanisms, with the potential translatability of key learnings from mouse models to the clinic. The common inbred mouse strain BALB/c is frequently used in immunological research. However, for the BALB/c histocompatibility (H)-2 alleles availability of conditional ligand has been limited. To overcome this challenge, we design and experimentally validate conditional ligands restricted to murine MHC class I alleles H2Dd and H2Kd. In addition, we demonstrate the ability of the three H2d molecules and two additional C57BL/6 H2b molecules folded in-house with conditional ligands to generate fluorescently labeled peptide-H2 tetramers that allow staining of antigen-specific CD8+ T cells in splenocyte samples. Finally, we generate large peptide-H-2 multimer libraries with a DNA-barcode labeling system for high-throughput interrogation of CD8+ T cell specificity in murine splenocyte samples. Consequently, the described techniques will contribute to our understanding of the antigen-specific CD8+ T cell repertoire in murine preclinical models of various diseases.
Collapse
Affiliation(s)
- Trine Sundebo Meldgaard
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
- Novo Nordisk, Copenhagen, Denmark
| | - Nadia Viborg
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
- Evaxion Biotech, Hørsholm, Denmark
| | - Sara Suarez Hernandez
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
- RIVM National Institute for Public Health and the Environment, Utrecht, The Netherlands
| | - Dario Vazquez Albacete
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
- Novonesis, Copenhagen, Denmark
| | - Tripti Tamhane
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark.
| | - Sine Reker Hadrup
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
2
|
Jiang N, Malone M, Chizari S. Antigen-specific and cross-reactive T cells in protection and disease. Immunol Rev 2023; 316:120-135. [PMID: 37209375 PMCID: PMC10524458 DOI: 10.1111/imr.13217] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/22/2023]
Abstract
Human T cells have a diverse T-cell receptor (TCR) repertoire that endows them with the ability to identify and defend against a broad spectrum of antigens. The universe of possible antigens that T cells may encounter, however, is even larger. To effectively surveil such a vast universe, the T-cell repertoire must adopt a high degree of cross-reactivity. Likewise, antigen-specific and cross-reactive T-cell responses play pivotal roles in both protective and pathological immune responses in numerous diseases. In this review, we explore the implications of these antigen-driven T-cell responses, with a particular focus on CD8+ T cells, using infection, neurodegeneration, and cancer as examples. We also summarize recent technological advances that facilitate high-throughput profiling of antigen-specific and cross-reactive T-cell responses experimentally, as well as computational biology approaches that predict these interactions.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, 19104
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, 19104
| | - Michael Malone
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| | - Shahab Chizari
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
3
|
Marrack P. Obsessive-Compulsive Behavior Isn't Necessarily a Bad Thing. Annu Rev Immunol 2020; 38:1-21. [PMID: 31594433 DOI: 10.1146/annurev-immunol-072319-033325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It is difficult to believe that in about 1960 practically nothing was known about the thymus and some of its products, T cells bearing αβ receptors for antigen. Thus I was lucky to join the field of T cell biology almost at its beginning, when knowledge about the cells was just getting off the ground and there was so much to discover. This article describes findings about these cells made by others and myself that led us all from ignorance, via complete confusion, to our current state of knowledge. I believe I was fortunate to practice science in very supportive institutions and with very collaborative colleagues in two countries that both encourage independent research by independent scientists, while simultaneously ignoring or somehow being able to avoid some of the difficulties of being a woman in what was, at the time, a male-dominated profession.
Collapse
Affiliation(s)
- Philippa Marrack
- Department of Biomedical Research, National Jewish Health, Denver, Colorado 80206, USA; .,Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
4
|
Rodriguez-Barbosa JI, Azuma M, Zelinskyy G, Perez-Simon JA, del Rio ML. Critical role of PD-L1 expression on non-tumor cells rather than on tumor cells for effective anti-PD-L1 immunotherapy in a transplantable mouse hematopoietic tumor model. Cancer Immunol Immunother 2020; 69:1001-1014. [DOI: 10.1007/s00262-020-02520-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 02/14/2020] [Indexed: 12/18/2022]
|
5
|
Hayakawa Y, Kawada M, Nishikawa H, Ochiya T, Saya H, Seimiya H, Yao R, Hayashi M, Kai C, Matsuda A, Naoe T, Ohtsu A, Okazaki T, Saji H, Sata M, Sugimura H, Sugiyama Y, Toi M, Irimura T. Report on the use of non-clinical studies in the regulatory evaluation of oncology drugs. Cancer Sci 2016; 107:189-202. [PMID: 26919617 PMCID: PMC4768389 DOI: 10.1111/cas.12857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 01/04/2023] Open
Abstract
Non-clinical studies are necessary at each stage of the development of oncology drugs. Many experimental cancer models have been developed to investigate carcinogenesis, cancer progression, metastasis, and other aspects in cancer biology and these models turned out to be useful in the efficacy evaluation and the safety prediction of oncology drugs. While the diversity and the degree of engagement in genetic changes in the initiation of cancer cell growth and progression are widely accepted, it has become increasingly clear that the roles of host cells, tissue microenvironment, and the immune system also play important roles in cancer. Therefore, the methods used to develop oncology drugs should continuously be revised based on the advances in our understanding of cancer. In this review, we extensively summarize the effective use of those models, their advantages and disadvantages, ranges to be evaluated and limitations of the models currently used for the development and for the evaluation of oncology drugs.
Collapse
Affiliation(s)
- Yoshihiro Hayakawa
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Division of Pathogenic BiochemistryDepartment of BioscienceInstitute of Natural MedicineUniversity of ToyamaToyamaJapan
| | - Manabu Kawada
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Institute of Microbial ChemistryMicrobial Chemistry Research FoundationNumazu‐shiJapan
| | - Hiroyoshi Nishikawa
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Division of Cancer ImmunologyExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterChibaJapan
| | - Takahiro Ochiya
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Division of Molecular and Cellular MedicineNational Cancer Center Research InstituteTokyoJapan
| | - Hideyuki Saya
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Division of Gene RegulationInstitute for Advanced Medical ResearchSchool of MedicineKeio UniversityTokyoJapan
| | - Hiroyuki Seimiya
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Division of Molecular BiotherapyCancer Chemotherapy CenterJapanese Foundation for Cancer ResearchTokyoJapan
| | - Ryoji Yao
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Division of Cell BiologyCancer InstituteJapanese Foundation for Cancer ResearchTokyoJapan
| | - Masahiro Hayashi
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Department of PharmacyToranomon HospitalTokyoJapan
| | - Chieko Kai
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Laboratory Animal Research CenterInstitute of Medical ScienceThe University of TokyoTokyoJapan
| | - Akira Matsuda
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Department of Medicinal ChemistryFaculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Tomoki Naoe
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- National Hospital Organization Nagoya Medical CenterNagoyaJapan
| | - Atsushi Ohtsu
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterChibaJapan
| | - Taku Okazaki
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Division of Immune RegulationInstitute for Genome ResearchTokushima UniversityTokushimaJapan
| | - Hideo Saji
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Department of Patho‐Functional Bioanalysis, Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
| | - Masataka Sata
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Department of Cardiovascular MedicineInstitute of Biomedical SciencesTokushima University Graduate SchoolTokushimaJapan
| | - Haruhiko Sugimura
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Department of Tumor PathologyHamamatsu University School of MedicineShizuokaJapan
| | - Yuichi Sugiyama
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Sugiyama LaboratoryRIKEN Innovation CenterRIKEN Cluster for Industry PartnershipsKanagawaJapan
| | - Masakazu Toi
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Department of Breast SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Tatsuro Irimura
- Subcommittee on Non‐clinical StudiesThe Science Board to the Pharmaceuticals and Medical Devices AgencyTokyoJapan
- Juntendo University School of MedicineTokyoJapan
| |
Collapse
|
6
|
Antón LC, Yewdell JW. Translating DRiPs: MHC class I immunosurveillance of pathogens and tumors. J Leukoc Biol 2014; 95:551-62. [PMID: 24532645 PMCID: PMC3958739 DOI: 10.1189/jlb.1113599] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/15/2014] [Accepted: 01/19/2014] [Indexed: 11/24/2022] Open
Abstract
MHC class I molecules display oligopeptides on the cell surface to enable T cell immunosurveillance of intracellular pathogens and tumors. Speed is of the essence in detecting viruses, which can complete a full replication cycle in just hours, whereas tumor detection is typically a finding-the-needle-in-the-haystack exercise. We review current evidence supporting a nonrandom, compartmentalized selection of peptidogenic substrates that focuses on rapidly degraded translation products as a main source of peptide precursors to optimize immunosurveillance of pathogens and tumors.
Collapse
Affiliation(s)
- Luis C Antón
- 1.NIAID, NIH, Bldg. 33, Bethesda, MD 20892, USA.
| | | |
Collapse
|
7
|
Wang Y, Takao Y, Harada M, Yutani S, Ide T, Sata M, Itoh K, Yamada A. New Epitope Peptides Derived from Hepatitis C Virus (HCV) 2a Which Have the Capacity to Induce Cytotoxic T Lymphocytes in HLA-A2+HCV-Infected Patients. Microbiol Immunol 2013; 50:857-65. [PMID: 17116980 DOI: 10.1111/j.1348-0421.2006.tb03861.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Because cytotoxic T lymphocytes (CTLs) play an important role in the specific immunotherapy of hepatitis C virus (HCV) infection, a series of CTL epitopes has been defined from HCV genotype 1a or 1b protein. Here, we attempted to identify HCV2a-derived epitopes that are capable of inducing HLA-A2-restricted and peptide-specific CTLs. Peripheral blood mononuclear cells (PBMCs) of HLA-A2+ HCV2ainfected patients or healthy donors were stimulated in vitro with each of the HCV2a-derived peptides, which were prepared based on the HLA-A2-binding motif, and their peptide-specific and HLA-A2-restricted cytotoxicities were examined. The HCV2a 432-441, HCV2a 716-724, and HCV2a 2251-2260 peptides were found to efficiently induce peptide-specific CTLs from the PBMCs of HLA-A2+ HCV2ainfected patients. Cytotoxicity was mainly mediated by CD8+ T cells in a HLA class I-restricted manner. These results indicate that the HCV2a 432-441, HCV2a 716-724, and HCV2a 2251-2260 peptides might be applicable for peptide-based immunotherapy of HLA-A2+ HCV2a-infected patients.
Collapse
Affiliation(s)
- Yi Wang
- Cancer Vaccine Development Division, Kurume University Research Center for Innovative Cancer Therapy, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Vaccine-elicited human T cells recognizing conserved protein regions inhibit HIV-1. Mol Ther 2013; 22:464-475. [PMID: 24166483 DOI: 10.1038/mt.2013.248] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/16/2013] [Indexed: 12/14/2022] Open
Abstract
Virus diversity and escape from immune responses are the biggest challenges to the development of an effective vaccine against HIV-1. We hypothesized that T-cell vaccines targeting the most conserved regions of the HIV-1 proteome, which are common to most variants and bear fitness costs when mutated, will generate effectors that efficiently recognize and kill virus-infected cells early enough after transmission to potentially impact on HIV-1 replication and will do so more efficiently than whole protein-based T-cell vaccines. Here, we describe the first-ever administration of conserved immunogen vaccines vectored using prime-boost regimens of DNA, simian adenovirus and modified vaccinia virus Ankara to uninfected UK volunteers. The vaccine induced high levels of effector T cells that recognized virus-infected autologous CD4(+) cells and inhibited HIV-1 replication by up to 5.79 log10. The virus inhibition was mediated by both Gag- and Pol- specific effector CD8(+) T cells targeting epitopes that are typically subdominant in natural infection. These results provide proof of concept for using a vaccine to target T cells at conserved epitopes, showing that these T cells can control HIV-1 replication in vitro.
Collapse
|
9
|
An optimized method for establishing high purity murine CD8+ T cell cultures. J Immunol Methods 2012; 387:173-80. [PMID: 23098837 DOI: 10.1016/j.jim.2012.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 09/19/2012] [Accepted: 10/16/2012] [Indexed: 02/06/2023]
Abstract
Establishing CD8(+) T cell cultures has been empirical and the published methods have been largely individual laboratory based. In this study, we optimized culturing conditions and show that IL-2 concentration is the most critical factor for the success of establishing CD8(+) T cell cultures. High IL-2 concentration encouraged T cells to non-specifically proliferate, express a B cell marker, B220, and undergo apoptosis. These cells also lose typical irregular T cell morphology and are incapable of sustaining long-term cultures. Using tetramer and intracellular cytokine assessments, we further demonstrated that many antigen-specific T cells have been rendered nonfunctional when expanded under high IL-2 concentration. When IL-2 is used in the correct range, B220-mediated cell depletion greatly enhanced the success rate of such T cell cultures.
Collapse
|
10
|
Jeon YH, Lee HW, Lee YL, Kim JE, Hwang MH, Jeong SY, Lee SW, Ahn BC, Ha JH, Lee J. Combined E7-dendritic cell-based immunotherapy and human sodium/iodide symporter radioiodine gene therapy with monitoring of antitumor effects by bioluminescent imaging in a mouse model of uterine cervical cancer. Cancer Biother Radiopharm 2011; 26:671-9. [PMID: 22091632 DOI: 10.1089/cbr.2011.1081] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Using a uterine cervical cancer cell line expressing human papillomavirus (HPV) 16 E7 antigen and bioluminescent imaging (BLI), we evaluated the therapeutic potential of combined immunotherapy using transfected dendritic cells (DC-E7) and human sodium/iodide symporter (hNIS) radioiodine gene therapy in a xenograft animal cancer model. Dendritic cells expressing either E7 antigen (DC-E7) or no-insert (DC-no insert) were made for immunization materials, and murine uterine cervical cancer cell line coexpressing E7, firefly luciferase, hNIS, and EGFP genes (TC-1/FNG) were prepared for the animal tumor model. C57BL/6 mice were divided into five therapy groups (phosphate-buffered saline [PBS], DC-no insert, DC-E7, I-131, and DC-E7+I-131 groups). Single therapy with either DC-E7 or I-131 induced greater retardation in tumor growth compared with PBS or DC-no insert groups, and it resulted in some tumor-free mice (DC-E7 and I-131 groups, 40% and 20%, respectively). Combination therapy with DC-E7 and I-131 dramatically inhibited tumor growth, thus causing complete disappearance of tumors in all mice, and these effects were further confirmed by BLI in vivo. In conclusion, complete disappearance of the tumor was achieved with combined DC-E7 vaccination and hNIS radioiodine gene therapy in a mouse model with E7-expressing uterine cervical cancer, and serial BLIs successfully demonstrated antitumor effects in vivo.
Collapse
Affiliation(s)
- Yong Hyun Jeon
- Department of Nuclear Medicine, Kyungpook National University, Daegu, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Jiang J, Fisher EM, Murasko DM. CD8 T cell responses to influenza virus infection in aged mice. Ageing Res Rev 2011; 10:422-7. [PMID: 21315186 DOI: 10.1016/j.arr.2011.02.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/28/2011] [Accepted: 02/01/2011] [Indexed: 01/15/2023]
Abstract
Influenza is one of the most common infectious diseases afflicting humans, particularly the elderly. The murine model has been widely employed for investigation of immunity to influenza virus infection. In this paper, we review the recent advances in understanding the diminished CD8 T cell immune response to influenza virus infection in aged mice. Possible mechanisms of impaired CD8 T cell responses with aging are addressed, including: (1) the role of dendritic cells (DCs); (2) the effect of age-associated changes in the T cell repertoire; and (3) the interactions with CD4 T cells, including T regulatory (Treg) cells and CD4 T helper cells. The aged murine model of the CD8 T cell response to influenza virus is helping to elucidate the mechanisms of immunosenescence which can lead to therapeutic improvements in the primary CD8 T cell response to new infections, as well as the development of new strategies for immunization to prevent influenza in the elderly.
Collapse
|
12
|
Lesterhuis WJ, de Vries IJM, Schreibelt G, Lambeck AJ, Aarntzen EH, Jacobs JF, Scharenborg NM, van de Rakt MW, de Boer AJ, Croockewit S, van Rossum MM, Mus R, Oyen WJ, Boerman OC, Lucas S, Adema GJ, Punt CJ, Figdor CG. Route of Administration Modulates the Induction of Dendritic Cell Vaccine–Induced Antigen-Specific T Cells in Advanced Melanoma Patients. Clin Cancer Res 2011; 17:5725-35. [DOI: 10.1158/1078-0432.ccr-11-1261] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
13
|
Reikie BA, Smolen KK, Fortuno ES, Loeffler DIM, Cai B, Blimkie D, Kollmann TR. A single immunization near birth elicits immediate and lifelong protective immunity. Vaccine 2010; 29:83-90. [PMID: 21034825 DOI: 10.1016/j.vaccine.2010.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 09/28/2010] [Accepted: 10/05/2010] [Indexed: 02/03/2023]
Abstract
Most existing vaccines do not induce protective immunity immediately following birth, nor do they retain protective efficacy in the latter years of life without booster doses. Using a mouse model, we present evidence that a live-replicating vaccine administered only once shortly after birth was able to induce both immediate and lifelong protection. Newborn mice immunized with a safe, highly attenuated strain of Listeria monocytogenes (Lm) were already protected by day 7 post-vaccination when challenged with a virulent strain of Lm. Furthermore, all mice remained fully protected for 2 years after only a single immunization. Vaccine-specific T cell immune responses were still detectable 2 years later, indicating long-lived immune memory even in neonatal vaccine recipients. Analysis of memory precursor subsets, specific for antigens homologous to Lm or a model vaccine (Ova), demonstrated remarkable similarity between adult and neonatal vaccine recipient effector and central memory CD8 T cell development. The magnitude of expansion of antigen specific memory T cells post-infectious challenge correlated with protection in both groups. This is the first direct evidence that vaccination--even in the absence of a booster dose--is capable of inducing immediate and lifelong protective immune memory regardless of age at the time of initial vaccination.
Collapse
Affiliation(s)
- Brian A Reikie
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, V5Z 1M9, Canada
| | | | | | | | | | | | | |
Collapse
|
14
|
Juang J, Ebert PJ, Feng D, Garcia KC, Krogsgaard M, Davis MM. Peptide-MHC heterodimers show that thymic positive selection requires a more restricted set of self-peptides than negative selection. J Exp Med 2010; 207:1223-34. [PMID: 20457759 PMCID: PMC2882826 DOI: 10.1084/jem.20092170] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 04/08/2010] [Indexed: 11/04/2022] Open
Abstract
T cell selection and maturation in the thymus depends on the interactions between T cell receptors (TCRs) and different self-peptide-major histocompatibility complex (pMHC) molecules. We show that the affinity of the OT-I TCR for its endogenous positively selecting ligands, Catnb-H-2Kb and Cappa1-H-2Kb, is significantly lower than for previously reported positively selecting altered peptide ligands. To understand how these extremely weak endogenous ligands produce signals in maturing thymocytes, we generated soluble monomeric and dimeric peptide-H-2Kb ligands. Soluble monomeric ovalbumin (OVA)-Kb molecules elicited no detectable signaling in OT-I thymocytes, whereas heterodimers of OVA-Kb paired with positively selecting or nonselecting endogenous peptides, but not an engineered null peptide, induced deletion. In contrast, dimer-induced positive selection was much more sensitive to the identity of the partner peptide. Catnb-Kb-Catnb-Kb homodimers, but not heterodimers of Catnb-Kb paired with a nonselecting peptide-Kb, induced positive selection, even though both ligands bind the OT-I TCR with detectable affinity. Thus, both positive and negative selection can be driven by dimeric but not monomeric ligands. In addition, positive selection has much more stringent requirements for the partner self-pMHC.
Collapse
Affiliation(s)
- Jeremy Juang
- The Department of Microbiology and Immunology, Department of Molecular and Cellular Physiology, Department of Structural Biology, and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| | - Peter J.R. Ebert
- The Department of Microbiology and Immunology, Department of Molecular and Cellular Physiology, Department of Structural Biology, and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| | - Dan Feng
- The Department of Microbiology and Immunology, Department of Molecular and Cellular Physiology, Department of Structural Biology, and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| | - K. Christopher Garcia
- The Department of Microbiology and Immunology, Department of Molecular and Cellular Physiology, Department of Structural Biology, and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| | - Michelle Krogsgaard
- Department of Pathology and New York University Cancer Institute, New York University School of Medicine, New York, NY 10016
| | - Mark M. Davis
- The Department of Microbiology and Immunology, Department of Molecular and Cellular Physiology, Department of Structural Biology, and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
15
|
Wischhusen J, Waschbisch A, Wiendl H. Immune-refractory cancers and their little helpers--an extended role for immunetolerogenic MHC molecules HLA-G and HLA-E? Semin Cancer Biol 2007; 17:459-68. [PMID: 17768067 DOI: 10.1016/j.semcancer.2007.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 07/04/2007] [Accepted: 07/19/2007] [Indexed: 11/18/2022]
Abstract
There is strong evidence to support a role for non-classical MHC class I (class Ib) molecules, most notably HLA-E and HLA-G in tumour immune escape. In this article, we summarize the current knowledge on their expression, regulation and functional relevance in various malignancies, particularly brain tumours. Special emphasis is devoted to the phenomenon that these tolerogenic molecules are expressed by non-transformed cells that are found in close neighborhood to tumour cells representing either parenchymal cells or immune cells attracted to the tumour microenvironment. Here they may act as "natural" or "inducible" suppressors of anti-tumoural immune responses. We thus speculate about the role of HLA-G expressing T cells, a novel population of natural regulatory cells that was identified recently. It is suggested that various cell types within a tumour cooperate in order to inhibit anti-tumour immunity-and that immunetolerogenic HLA-G may play a major role in this context.
Collapse
Affiliation(s)
- Jörg Wischhusen
- Department for Obstetrics and Gynecology, Julius-Maximilians-University Würzburg, School of Medicine, Josef-Schneider-Strasse 4, 97080 Würzburg, Germany
| | | | | |
Collapse
|
16
|
Disis ML, Bernhard H, Gralow JR, Hand SL, Emery SR, Calenoff E, Cheever MA. Immunity to the HER-2/neu oncogenic protein. CIBA FOUNDATION SYMPOSIUM 2007; 187:198-207; discussion 207-11. [PMID: 7540970 DOI: 10.1002/9780470514672.ch13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The study of oncogenic viruses led to the discovery that transforming retroviruses contain oncogenes homologous with and/or derived from cellular proto-oncogenes. In humans malignant transformation is often the result of the activation of proto-oncogenes. Normal proto-oncogenes can be activated to transforming proto-oncogenes by a variety of mechanisms including point mutation, translocation and amplification. Development of successful strategies for the immunotherapy of human cancers is an area of intense investigation. Part of the problem in developing cancer-specific immunotherapy has been the lack of well-defined tumour antigens. Our laboratory has focused on the question of whether oncogenic proteins expressed by transforming proto-oncogenes can serve as targets for immune attack. Some patients with HER-2/Neu-positive breast cancer have an existent immune response to the HER-2/neu protein with no clinical signs of autoimmunity, supporting the idea that overexpressed oncogenic proteins can be targeted in therapy without fear of destructive autoimmunity. The identification of candidate cytotoxic T lymphocyte epitopes might allow the generation of tumour-specific cytotoxic T lymphocytes for use in therapy and identify potential epitopes for peptide vaccines.
Collapse
Affiliation(s)
- M L Disis
- Department of Medicine, University of Washington, Seattle 98195, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Purtha WE, Myers N, Mitaksov V, Sitati E, Connolly J, Fremont DH, Hansen TH, Diamond MS. Antigen-specific cytotoxic T lymphocytes protect against lethal West Nile virus encephalitis. Eur J Immunol 2007; 37:1845-54. [PMID: 17559174 DOI: 10.1002/eji.200737192] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Infection with West Nile virus (WNV) causes fatal encephalitis in immunocompromised animals. Previous studies in mice have established that T cell protection is required for clearance of WNV infection from tissues and preventing viral persistence. The current study assessed whether specific WNV peptide epitopes could elicit a cytotoxic T lymphocyte (CTL) response capable of protecting against virus infection. Hidden Markov model analysis was used to identify WNV-encoded peptides that bound the MHC class I proteins K(b) or D(b). Of the 35 peptides predicted to bind MHC class I molecules, one immunodominant CTL recognition peptide was identified in each of the envelope and non-structural protein 4B genes. Addition of these but not control peptides to CD8(+) T cells from WNV-infected mice induced IFN-gamma production. CTL clones that were generated ex vivo lysed peptide-pulsed or WNV-infected target cells in an antigen-specific manner. Finally, adoptive transfer of a mixture of envelope- and non-structural protein 4B-specific CTL to recipient mice protected against lethal WNV challenge. Based on this, we conclude that CTL responses against immundominant WNV epitopes confer protective immunity and thus should be targets for inclusion in new vaccines.
Collapse
Affiliation(s)
- Whitney E Purtha
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Sengupta S, Jayaraman P, Chilton PM, Casella CR, Mitchell TC. Unrestrained glycogen synthase kinase-3 beta activity leads to activated T cell death and can be inhibited by natural adjuvant. THE JOURNAL OF IMMUNOLOGY 2007; 178:6083-91. [PMID: 17475833 DOI: 10.4049/jimmunol.178.10.6083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Activated T cell death (ATCD) after peak clonal expansion is required for effective homeostasis of the immune system. Using a mouse model of T cell clonal expansion and contraction, we found that regulation of the proapoptotic kinase glycogen synthase kinase (GSK)-3beta plays a decisive role in determining the extent to which T cells are eliminated after activation. Involvement of GSK-3beta in ATCD was tested by measuring T cell survival after GSK-3beta inhibition, either ex vivo with chemical and pharmacological inhibitors or in vivo by retroviral expression of a dominant-negative form of GSK-3. We also measured amounts of inactivating phosphorylation of GSK-3beta (Ser9) in T cells primed in the presence or absence of LPS. Our results show that GSK-3beta activity is required for ATCD and that its inhibition promoted T cell survival. Adjuvant treatment in vivo maintained GSK-3beta (Ser9) phosphorylation in activated T cells, whereas with adjuvant-free stimulation it peaked and then decayed as the cells became susceptible to ATCD. We conclude that the duration of GSK-3beta inactivation determines activated T cell survival and that natural adjuvant stimulation decreases the severity of clonal contraction in part by keeping a critical proapoptotic regulatory factor, GSK-3beta, inactivated.
Collapse
Affiliation(s)
- Sadhak Sengupta
- Institute for Cellular Therapeutics and Department of Microbiology and Immunology, University of Louisville School of Medicine, KY 40202, USA
| | | | | | | | | |
Collapse
|
19
|
Keating R, Yue W, Rutigliano JA, So J, Olivas E, Thomas PG, Doherty PC. Virus-specific CD8+ T cells in the liver: armed and ready to kill. THE JOURNAL OF IMMUNOLOGY 2007; 178:2737-45. [PMID: 17312116 DOI: 10.4049/jimmunol.178.5.2737] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Influenza A virus infection of C57BL/6 mice is a well-characterized model for studying CD8+ T cell-mediated immunity. Analysis of primary and secondary responses showed that the liver is highly enriched for CD8+ T cells specific for the immunodominant H2D(b)NP(366-374) (D(b)NP(366)) epitope. Functional analysis established that these liver-derived virus-specific CD8+ T cells are fully competent cytotoxic effectors and IFN-gamma secretors. In addition, flow cytometric analysis of early apoptotic cells showed that these influenza-specific CD8+ T cells from liver are as viable as those in the spleen, bronchoalveolar lavage, mediastinal lymph nodes, or lung. Moreover, cytokine profiles of the influenza-specific CD8+ T cells recovered from different sites were consistent with the bronchoalveolar lavage, rather than liver population, being the most susceptible to activation-induced cell death. Importantly, adoptively transferred influenza virus-specific CD8+ T cells from the liver survived and were readily recalled after virus challenge. Together, these results show clearly that the liver is not a "graveyard" for influenza virus-specific CD8+ T cells.
Collapse
Affiliation(s)
- Rachael Keating
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Santegoets SJAM, Schreurs MWJ, Masterson AJ, Liu YP, Goletz S, Baumeister H, Kueter EWM, Lougheed SM, van den Eertwegh AJM, Scheper RJ, Hooijberg E, de Gruijl TD. In vitro priming of tumor-specific cytotoxic T lymphocytes using allogeneic dendritic cells derived from the human MUTZ-3 cell line. Cancer Immunol Immunother 2006; 55:1480-90. [PMID: 16468034 PMCID: PMC11030798 DOI: 10.1007/s00262-006-0142-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Accepted: 01/24/2006] [Indexed: 12/16/2022]
Abstract
The adoptive transfer of in vitro-induced and expanded tumor-specific cytotoxic T lymphocytes (CTL) presents a promising immunotherapeutic approach for the treatment of cancer. The in vitro induction of tumor-reactive CTL requires repeated stimulation of CTL precursors with dendritic cells (DC). To circumvent problems like scarcity of blood DC precursors and donor variability, it would be attractive to use DC from a non-autologous, unlimited source. DCs derived from the human acute myeloid leukemia (AML) cell line MUTZ-3 are attractive candidates since these DCs closely resemble monocyte-derived DC (MoDC) in terms of phenotype and T cell stimulatory capacity. Here we demonstrate that functional CTL clones could be generated against multiple tumor-associated antigens, i.e., human telomerase reverse transcriptase (hTERT), ErbB3-binding protein-1 (Ebp1), carcinoembryonic antigen (CEA) and Her-2/neu, by stimulating CD8beta(+) CTL precursors with peptide-loaded allogeneic, HLA-A2-matched MUTZ-3-derived DC. A consistent induction capacity, as determined by MHC tetramer-binding, was found in multiple donors and comparable to autologous peptide-loaded MoDC. Functional characterization at the clonal level revealed the priming of CTL that recognized endogenously processed epitopes on tumor cell lines in an HLA-A2-restricted fashion. Our data indicate that MUTZ-3-derived DC can be used as stimulator cells for in vitro priming and expansion of functional TAA-specific effector CTL. MUTZ-3-derived DCs thus represent a ready and standardized source of allogeneic DC to generate CTL for therapeutic adoptive transfer strategies.
Collapse
Affiliation(s)
- Saskia J. A. M. Santegoets
- Departments of Pathology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| | - Marco W. J. Schreurs
- Departments of Pathology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| | - Allan J. Masterson
- Medical Oncology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| | - Ying Poi Liu
- Departments of Pathology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| | - Steffen Goletz
- NEMOD Biotherapeutics, GmbH & Co. KG, Berlin, Germany
- GLYCOTOPE GmbH, 13125 Berlin, Germany
| | | | - Esther W. M. Kueter
- Departments of Pathology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| | - Sinéad M. Lougheed
- Medical Oncology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| | | | - Rik J. Scheper
- Departments of Pathology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
- Present Address: De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Erik Hooijberg
- Departments of Pathology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| | - Tanja D. de Gruijl
- Medical Oncology, VU University Medical Center, P.O. box 7057, 1007 MB Amsterdam, The Netherlands
| |
Collapse
|
21
|
Saha A, Chatterjee SK, Foon KA, Bhattacharya-Chatterjee M. Anti-idiotype antibody induced cellular immunity in mice transgenic for human carcinoembryonic antigen. Immunology 2006; 118:483-96. [PMID: 16895556 PMCID: PMC1782317 DOI: 10.1111/j.1365-2567.2006.02391.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In the present study, we have analysed the detailed cellular immune mechanisms involved in tumour rejection in carcinoembryonic antigen (CEA) transgenic mice after immunization with dendritic cells (DC) pulsed with an anti-idiotype (Id) antibody, 3H1, which mimics CEA. 3H1-pulsed DC vaccinations resulted in induction of CEA specific cytotoxic T lymphocyte (CTL) responses in vitro and the rejection of CEA-transfected MC-38 murine colon carcinoma cells, C15, in vivo (Saha et al.,Cancer Res 2004; 64: 4995-5003). These CTL mediated major histocompatibility complex (MHC) class I-restricted tumour cell lysis, production of interferon-gamma (IFN-gamma) and tumour necrosis factor-alpha (TNF-alpha), and expression of Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL) in response to C15 cells. CTL used perforin-, FasL-, and TRAIL-mediated death pathways to lyse C15 cells, although perforin-mediated killing was the predominant lytic mechanism in vitro. The cytokines IFN-gamma and TNF-alpha synergistically enhanced surface expression of Fas, TRAIL receptor, MHC class I and class II on C15 cells that increased the sensitivity of tumour cells to CTL lysis. CTL activity generated in 3H1-pulsed DC immunized mice was directed against an epitope defined by the idio-peptide LCD-2, derived from 3H1. In vivo lymphocyte depletion experiments demonstrated that induction of CTL response and antitumour immunity was dependent on both CD4+ and CD8+ T cells. The analysis of splenocytes of immunized mice that had rejected C15 tumour growth revealed up-regulated surface expression of memory phenotype Ly-6C and CD44 on both CD4+ and CD8+ T cells. The adoptive transfer experiments also suggested the role of both CD4+ and CD8+ T cells in this model system. Furthermore, mice that had rejected C15 tumour growth, developed tumour-specific immunological memory.
Collapse
Affiliation(s)
- Asim Saha
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH45267, USA
| | | | | | | |
Collapse
|
22
|
Fong CL, Mok CL, Hui KM. Intramuscular immunization with plasmid coexpressing tumour antigen and Flt-3L results in potent tumour regression. Gene Ther 2006; 13:245-56. [PMID: 16163376 DOI: 10.1038/sj.gt.3302639] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DC) are professional antigen-presenting cells capable of initiating a potent primary immune response, making them an attractive target for cancer immunotherapy. Flt-3 ligand (Flt-3L) is a haematopoietic growth factor that efficiently induces DC expansion in vivo. To achieve a more efficient and effective method of priming tumour-specific, DC-mediated immune response, we generated a DNA vaccine comprising both human Flt-3L and the tumour antigen, MUC-1 (pNGVL-hFLex-MUC-1). We report that pNGVL-hFLex-MUC-1 is able to induce antigen-specific CTL immunity in vivo, resulting in a potent anti-tumour response, and that the Flt-3L component is essential to the efficacy of the DNA vaccine. Moreover, the route of immunization is critical in determining the type of immune response generated; intramuscular (i.m.) immunization with pNGVL-hFLex-MUC-1 conferred tumour protection in contrast to poor response with hydrodynamic-based intravenous delivery. Post-i.m. immunization, we observed a massive infiltration of mononuclear cells to the injection site, comprised predominantly of CD11c(+)/CD8alpha(-) DC. Therefore, we propose that Flt-3L acts as an adjuvant to recruit DC, thereby priming the anti-tumour response. However, systemic expansion of DC prior to immunization did not enhance the specific cellular response, suggesting that it is in situ recruitment or expansion of DC that is critical for pNGVL-hFLex-MUC-1 potency.
Collapse
Affiliation(s)
- C L Fong
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore
| | | | | |
Collapse
|
23
|
Christopher K, Liang Y, Mueller TF, DeFina R, He H, Haley KJ, Exley MA, Finn PW, Perkins DL. Analysis of the Major Histocompatibility Complex in Graft Rejection Revisited by Gene Expression Profiles. Transplantation 2004; 78:788-98. [PMID: 15385796 DOI: 10.1097/01.tp.0000128626.13712.88] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND The precise role of major histocompatibility complex (MHC) molecules in graft rejection remains incompletely understood. The important role of foreign peptides in the alloimmune response was recently recognized. METHODS We performed a comparative study of the functions of minor antigens Class I, Class II, and CD1 in murine cardiac allograft rejection by investigating the expression of a large panel of immune and inflammatory genes. To investigate the role of MHC Class II and I, our protocol analyzed allograft recipients deficient in MHC Class II and b2 microglobulin (b2-M), a critical component of the Class I heterodimer. We also included CD1 deficient recipients to differentiate effects in the beta2-M deficient strain due to CD1 deficiency versus the combined inactivation of CD1 and Class I. The serum cytokines tumor necrosis factor (TNF)-alpha interleukin (IL)-6, interferon (IFN)-gamma and IL-1beta were evaluated posttransplant by ELISA. The intragraft expression of 55 chemokines, chemokine receptors, and CD markers were measured by ribonuclease protection assay. The data were analyzed through hierarchical clustering dendrograms and self-organizing maps. RESULTS The analysis indicates that each gene deficiency induces both the upregulation and the downregulation of distinct subsets of genes and that similar kinetics of rejection can be attributed to different molecular mechanisms. CONCLUSIONS The study provides novel insights into the role of classical and non-classical MHC molecules in graft rejection.
Collapse
Affiliation(s)
- Kenneth Christopher
- Laboratory of Molecular Immunology, Renal Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lu J, Higashimoto Y, Appella E, Celis E. Multiepitope Trojan antigen peptide vaccines for the induction of antitumor CTL and Th immune responses. THE JOURNAL OF IMMUNOLOGY 2004; 172:4575-82. [PMID: 15034075 DOI: 10.4049/jimmunol.172.7.4575] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We describe in this study a strategy to produce synthetic vaccines based on a single polypeptide capable of eliciting strong immune responses to a combination CTL and Th epitopes with the purpose of treating malignancies or preventing infectious diseases. This strategy is based on the capacity of Trojan Ags to deliver exogenous Ags into the intracellular compartments, where processing into MHC-binding peptides takes place. Our previous work demonstrated that Trojan Ags containing a CTL epitope localized to intracellular compartments, where MHC class I-binding peptides were generated in a TAP-independent fashion by the action of various exopeptidases and the endopeptidase furin. In this study, we report that Trojan Ags containing several CTL epitopes joined via furin-sensitive linkers generated all of the corresponding MHC class I-binding peptides, which were recognized by CTL. However, Trojan Ags prepared with furin-resistant linkers failed to produce the MHC class I-binding peptides. We also present data indicating that Trojan Ags bearing both CTL and Th epitopes can generate the corresponding MHC class I- and II-binding peptides, which are capable of stimulating T cell responses. Most significantly, in vivo vaccination of mice with a single injection of multiepitope Trojan Ags resulted in strong CTL and Th responses that translated into significant antitumor responses in a model of malignant melanoma. The overall results indicate that Trojan Ags prepared with furin-sensitive linkers are ideal candidates for producing synthetic multiepitope vaccines for the induction of CTL and Th responses that could be used against a variety of diseases, including cancer.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Antigen-Presenting Cells/enzymology
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic/immunology
- Egg Proteins/immunology
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Female
- Furin/pharmacology
- Injections, Subcutaneous
- Mastocytoma/immunology
- Mastocytoma/prevention & control
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation
- Ovalbumin/immunology
- Peptide Fragments/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Helper-Inducer/immunology
- Transfection
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Jun Lu
- Department of Immunology and Cancer Center, Mayo College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
25
|
Staege MS, Gisch K, Reske-Kunz AB. Cytotoxic T cells with reciprocal antigenic peptide presentation function are not generally resistant to mutual lysis. Immunol Cell Biol 2003; 81:266-74. [PMID: 12848847 DOI: 10.1046/j.1440-1711.2003.t01-1-01164.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cytotoxic T cells normally express major histocompatibility complex class I molecules, to which their T cell antigen receptors are restricted. Therefore, a single cytotoxic T cell can not only act as a cytolytic effector cell, but also as an antigen-presenting cell for other cytotoxic T cells of the same or a different clone. In the present paper, we used a murine cytotoxic T cell clone, 10BK.1, recognizing the ovalbumin-derived peptide OVA257-264 in combination with H-2Kb to investigate the consequences of reciprocal antigen presentation by these cytotoxic T cells. These cells proliferate after incubation with the relevant peptide in the absence of added accessory cells, indicating reciprocal antigenic peptide presentation by the cytotoxic T cell. We found that reciprocal lysis of these cells was dependent on the time point of incubation with antigen. We did not observe reciprocal lysis of cytotoxic T cells used 30 days after the last restimulation with antigen. In contrast, 10BK.1 cells used two days after the last restimulation showed an increased capacity for reciprocal lysis. The lytic capacity decreased with time after restimulation. Reciprocal lysis of 10BK.1 cells depended on reciprocal peptide presentation by at least two 10BK.1 cells. Recognition of the antigenic peptide, together with class I molecules on the surface of classical syngeneic target cells did not induce lysis of freshly stimulated 10BK.1 cells, suggesting that reciprocal lysis was not just a consequence of re-activation of the cytotoxic T cells. Reciprocal destruction of freshly activated 10BK.1 cells proceeded independent of CD95/CD95 ligand. Despite an increased secretion of tumour necrosis factor-alpha by 10BK.1 cells on day 2 after antigen stimulation, compared with cells on day 30 after stimulation, tumour necrosis factor-alpha was not responsible for the reciprocal destruction of freshly stimulated 10BK.1 cells. Lysis of preactivated 10BK.1 cells was independent of autocrine interleukin-2 production by the cytotoxic T cells, but interleukin-2 was required for optimal priming of cytotoxic T cells for reciprocal lysis.
Collapse
Affiliation(s)
- Martin S Staege
- Clinical Research Unit Allergology, Department of Dermatology, Johannes Gutenberg University, Mainz, Germany.
| | | | | |
Collapse
|
26
|
Fong CL, Hui KM. Generation of potent and specific cellular immune responses via in vivo stimulation of dendritic cells by pNGVL3-hFLex plasmid DNA and immunogenic peptides. Gene Ther 2002; 9:1127-38. [PMID: 12170376 DOI: 10.1038/sj.gt.3301783] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2001] [Accepted: 04/07/2002] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DC) are the most potent professional antigen-presenting cells with exquisite capacity to interact with T cells to initiate strong primary cellular immune responses. The antigen-presenting capability of DC makes them attractive vehicles for the delivery of therapeutic cancer vaccines. Recently, we have demonstrated that the introduction of a recombinant gene encoding the human Flt3L gene into mice could result in the expansion of the DC population in vivo. In this report, we have introduced the human Flt-3L gene via naked DNA-based immunization in combination with the muc-1 tumor peptide to immunize mice. We demonstrated that the population of DC expanded following stimulation with the human Flt-3L gene in vivo is functional and they are able to elicit potent muc-1 peptide-specific cellular responses. The strategy described here allows the efficient generation of antigen-specific CTL immunity in vivo and has the potential to be applied in developing efficient protocols for antigen-specific immunotherapy of human malignancies.
Collapse
Affiliation(s)
- C L Fong
- Gene Vector Laboratory, Division of Cellular and Molecular Research, National Cancer Center, Singapore
| | | |
Collapse
|
27
|
Venturini S, Mosier DE, Burton DR, Poignard P. Characterization of human immunodeficiency virus type 1 (HIV-1) Gag- and Gag peptide-specific CD4(+) T-cell clones from an HIV-1-seronegative donor following in vitro immunization. J Virol 2002; 76:6987-99. [PMID: 12072499 PMCID: PMC136330 DOI: 10.1128/jvi.76.14.6987-6999.2002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Substantial evidence argues that human immunodeficiency virus type 1 (HIV-1)-specific CD4(+) T cells play an important role in the control of HIV-1 replication in infected individuals. Moreover, it is increasingly clear that an HIV vaccine should elicit potent cytotoxic lymphocyte and antibody responses that will likely require an efficient CD4(+) T-cell response. Therefore, understanding and characterizing HIV-specific CD4(+) T-cell responses is an important aim. Here we describe the generation of HIV-1 Gag- and Gag peptide-specific CD4(+) T-cell clones from an HIV-1-seronegative donor by in vitro immunization with HIV-1 Gag peptides. The Gag peptides were able to induce a strong CD4(+) T-cell immune response in peripheral blood mononuclear cells from the HIV-1-seronegative donor. Six Gag peptide-specific CD4(+) T-cell clones were isolated and their epitopes were mapped. The region of p24 between amino acids 201 and 300 of Gag was defined as the immunodominant region of Gag. A new T helper epitope in the p6 protein of Gag was identified. Two clones were shown to recognize Gag peptides and processed Gag protein, while the other four clones reacted only to Gag peptides under the experimental conditions used. Functional analysis of the clones indicated that both Th1 and Th2 types of CD4(+) T cells were obtained. One clone showed direct antigen-specific cytotoxic activity. These clones represent a valuable tool for understanding the cellular immune response to HIV-1, and the study provides new insights into the HIV-1-specific CD4(+) T-cell response and the induction of an anti-Gag and -Gag peptide cellular primary immune response in vitro.
Collapse
Affiliation(s)
- Sara Venturini
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
28
|
Micheletti F, Monini P, Fortini C, Rimessi P, Bazzaro M, Andreoni M, Giuliani M, Traniello S, Ensoli B, Gavioli R. Identification of cytotoxic T lymphocyte epitopes of human herpesvirus 8. Immunology 2002; 106:395-403. [PMID: 12100728 PMCID: PMC1782731 DOI: 10.1046/j.1365-2567.2002.01424.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human herpesvirus 8 (HHV-8) is a human gamma2-herpesvirus that is implicated in the development of Kaposi's sarcoma (KS), primary effusion lymphoma and Castelman's disease. Since the responses of cytotoxic T lymphocytes (CTL) play a key role in the control of herpesvirus infection, it is important to identify and to characterize the CTL target epitopes of HHV-8 viral antigens. In this study, using peptide-binding motifs, we selected potential human leucocyte antigen (HLA)-A2-binding peptides from kaposin A and glycoprotein H (gH), that are latent and lytic HHV-8 antigens, respectively. HLA-A2-binding peptides were tested for their capacity to induce CTL responses in HHV-8-negative healthy donors. By this approach, we found that the majority of individuals responded to two HHV-8-derived CTL epitopes, namely, VLLNGWRWRL (amino acids 16-25), which derives from kaposin A, and FLNWQNLLNV (amino acids 59-68), which derives from gH. In addition, memory CTL responses to these epitopes were detected in disease-free individuals infected by HHV-8 demonstrating that the two epitopes are relevant targets of CTL-mediated immunity in vivo. The identified epitopes may be investigated for the development of immunotherapeutic strategies against HHV-8-associated malignancies.
Collapse
Affiliation(s)
- Fabiola Micheletti
- Department of Biochemistry and Molecular Biology, University of Ferrara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Iyoda T, Shimoyama S, Liu K, Omatsu Y, Akiyama Y, Maeda Y, Takahara K, Steinman RM, Inaba K. The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo. J Exp Med 2002; 195:1289-302. [PMID: 12021309 PMCID: PMC2193756 DOI: 10.1084/jem.20020161] [Citation(s) in RCA: 498] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Dendritic cells (DCs) are able in tissue culture to phagocytose and present antigens derived from infected, malignant, and allogeneic cells. Here we show directly that DCs in situ take up these types of cells after fluorescent labeling with carboxyfluorescein succinimidyl ester (CFSE) and injection into mice. The injected cells include syngeneic splenocytes and tumor cell lines, induced to undergo apoptosis ex vivo by exposure to osmotic shock, and allogeneic B cells killed by NK cells in situ. The CFSE-labeled cells in each case are actively endocytosed by DCs in vivo, but only the CD8+ subset. After uptake, all of the phagocytic CD8+ DCs can form major histocompatibility complex class II-peptide complexes, as detected with a monoclonal antibody specific for these complexes. The CD8+ DCs also selectively present cell-associated antigens to both CD4+ and CD8+ T cells. Similar events take place with cultured DCs; CD8+ DCs again selectively take up and present dying cells. In contrast, both CD8+ and CD8- DCs phagocytose latex particles in culture, and both DC subsets present soluble ovalbumin captured in vivo. Therefore CD8+ DCs are specialized to capture dying cells, and this helps to explain their selective ability to cross present cellular antigens to both CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Tomonori Iyoda
- Department of Animal Development and Physiology, Graduate School of Biostudies. Department of Zoology, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sugawa S, Palliser D, Eisen HN, Chen J. How do cultured CD8(+) murine T cell clones survive repeated ligation of the TCR? Int Immunol 2002; 14:23-30. [PMID: 11751748 DOI: 10.1093/intimm/14.1.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Many murine T cell clones grow continuously in culture despite weekly ligation of their TCR by antigen. To learn how the cultured cells avoid or minimize antigen-induced cell death (AICD), we compared Fas and tumor necrosis factor (TNF) receptors (TNFR) on several long-term cultured CD8(+) T cell clones with those on naive and activated naive cells expressing the same TCR (2C). In contrast to the naive cells, Fas was absent on the cultured clones and the TNFR-II receptor, present initially at high levels on the cultured cells, was rapidly down-modulated in response to TCR ligation and had virtually disappeared by 2 h, when only approximately 10% of the cloned cells had been induced to express TNF-alpha. The extent of AICD of the cultured clones in response to cognate peptide-MHC on the presenting cells used for routine stimulation of the cultures was also considerably less than the massive cell death of the clones following exposure to anti-CD3 antibody plate-bound at high density.
Collapse
Affiliation(s)
- Satoshi Sugawa
- Center for Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | |
Collapse
|
31
|
Engler OB, Dai WJ, Sette A, Hunziker IP, Reichen J, Pichler WJ, Cerny A. Peptide vaccines against hepatitis B virus: from animal model to human studies. Mol Immunol 2001; 38:457-65. [PMID: 11741695 DOI: 10.1016/s0161-5890(01)00081-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
An estimated 400 million people are chronically infected with the hepatitis B virus (HBV). Chronic viral hepatitis infection incurs serious sequelae such as liver cirrhosis and hepatocellular carcinoma. Prevention and treatment, thus, represent an important target for public health. Preventive vaccines using HBsAg alone or combined with other antigens allow for the generation of neutralizing antibodies which effectively prevent infection in immunocompetent individuals. Cell-mediated immunological mechanisms are thought to be crucial in determining viral persistence or viral elimination. Therapeutic approaches aiming to shift cellular immunity towards viral elimination have been on the research agenda for many years. This paper summarizes pre-clinical and clinical results obtained with the use of immunogenic peptides formulated as vaccines to selectively boost cellular immune responses. Such vaccines are capable of generating cellular immune responses in animal models as well as in humans and represent an important step towards the development of a therapeutic vaccine against chronic hepatitis.
Collapse
Affiliation(s)
- O B Engler
- Clinic of Rheumatology and Clinical Immunology/Allergology, University Hospital, Inselspital Bern, 3010, Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
32
|
Yoshida A, Nagata T, Uchijima M, Koide Y. Protective CTL response is induced in the absence of CD4+ T cells and IFN-gamma by gene gun DNA vaccination with a minigene encoding a CTL epitope of Listeria monocytogenes. Vaccine 2001; 19:4297-306. [PMID: 11457557 DOI: 10.1016/s0264-410x(01)00146-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Our work was undertaken to learn the mechanism of induction of protective cytotoxic T lymphocytes (CTL) by gene gun DNA vaccination with p91m encoding an H-2Kd-restricted T cell epitope of listeriolysin O (LLO). Vaccination with p91m induced vigorous antigen-specific CD8+ CTL that produce IFN-gamma and was able to confer partial protection against listerial challenge. However, the p91m-induced protective immunity was revealed to be independent of the IFN-gamma and CD4+ T cell help. The CTL induction is also suggested to require neither adjuvant activity of the plasmid used nor IFN-gamma. The data may be feasible for the design of CTL inducing vaccines in various immunodeficiencies.
Collapse
Affiliation(s)
- A Yoshida
- Department of Microbiology and Immunology, Hamamatsu University School of Medicine, 431-3192, Hamamatsu, Japan
| | | | | | | |
Collapse
|
33
|
Knutson KL, Disis ML. Expansion of HER2/neu-specific T cells ex vivo following immunization with a HER2/neu peptide-based vaccine. Clin Breast Cancer 2001; 2:73-9. [PMID: 11899386 DOI: 10.3816/cbc.2001.n.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The identification and characterization of tumor antigens has facilitated the development of immune-based cancer prophylaxis and therapy. Cancer vaccines, like viral vaccines, may be effective in cancer prevention. Adoptive T-cell therapy, in contrast, may be more efficacious for the eradication of existing malignancies. Our group is examining the feasibility of antigen-specific adoptive T-cell therapy for the treatment of established cancer in the HER2/neu model. Transgenic mice overexpressing rat neu in mammary tissue develop malignancy, histologically similar to human HER2/neu-overexpressing breast cancer. These mice can be effectively immunized against a challenge with neu-positive tumor cells. Adoptive transfer of neu-specific T cells into tumor-bearing mice eradicates malignancy. Effective T-cell therapy relies on optimization of the ex vivo expansion of antigen-specific T cells. Two important elements of ex vivo antigen-specific T-cell growth that have been identified are (1) the preexisting levels of antigen-specific T cells and (2) the cytokine milieu used during ex vivo expansion of the T cells. Phase I clinical trials of HER2/neu-based peptide vaccination in human cancer patients have demonstrated that increased levels of HER2/neu-specific T-cells can be elicited after active immunization. Initiating cultures with greater numbers of antigen-specific T cells facilitates expansion. In addition, cytokines, such as interleukin-12, when added during ex vivo culturing along with interleukin-2 can selectively expand antigen-specific T-cells. Interleukin-12 also enhances antigen-specific functional measurements such as interferon-gamma and tumor necrosis factor-alpha release. Refinements in ex vivo expansion techniques may greatly improve the feasibility of tumor-antigen T-cell-based therapy for the treatment of advanced-stage HER2/neu-overexpressing breast malignancy.
Collapse
Affiliation(s)
- K L Knutson
- Division of Oncology, University of Washington, 1959 NE Pacific Street, HSB BB1321, Box 356527, Seattle, WA 98195-6527, USA.
| | | |
Collapse
|
34
|
Tolstrup AB, Duch M, Dalum I, Pedersen FS, Mouritsen S. Functional screening of a retroviral peptide library for MHC class I presentation. Gene 2001; 263:77-84. [PMID: 11223245 DOI: 10.1016/s0378-1119(00)00572-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We have used retroviral vector technology to develop a method for functional screening of combinatorial peptide libraries expressed inside mammalian cells with the ultimate goal of identifying new drug targets. The method was validated in a library screening experiment based on antigen presentation of small peptides. A library encoding SIXNXEKX-peptides, where X designates randomised positions corresponding to major histocompatibility (MHC) class I anchor residues, was generated in a retroviral vector. The library was transduced into a population of antigen presenting cells (APCs) known to mediate MHC class I restricted presentation of the SIINFEKL peptide. The cellular library was screened by using an antigen presentation assay in which a T cell hybridoma recognising the MHC class I/SIINFEKL peptide complex was employed. Using this experimental model, we identified two positive cellular clones both encoding SIINFEKL peptides with identical codon usage. This number corresponded well to the expected frequency of SIINFEKL in the library. The lack of identification of other peptides capable of activating the T-hybridoma supports previous findings of a high degree of specificity at the level of peptide-loading of MHC-molecules. The result further demonstrates the potential of using combinatorial libraries for functional screening and selection of effector peptides stably expressed in mammalian cells.
Collapse
Affiliation(s)
- A B Tolstrup
- M&E Biotech A/S, Kogle Allé 6, 2970, Hørsholm, Denmark.
| | | | | | | | | |
Collapse
|
35
|
Hegde NR, Srikumaran S. Reverse immunogenetic and polyepitopic approaches for the induction of cell-mediated immunity against bovine viral pathogens. Anim Health Res Rev 2000; 1:103-18. [PMID: 11708596 DOI: 10.1017/s1466252300000098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The control of several infectious diseases of animals by vaccination is perhaps the most outstanding accomplishment of veterinary medicine in the last century. Even the eradication of some pathogens is in sight, at least in some parts of the world. However, infectious diseases continue to cost millions of dollars to the livestock industry. One of the reasons for the failure to control certain pathogens is the limited emphasis placed on cell-mediated immunity (CMI) in the design of vaccines against these pathogens. Traditionally, vaccine-induced immunity has been studied in relation to antibody-mediated protection. More recent studies, however, have focused on understanding CMI and developing means of inducing CMI. This review focuses on recent advances made in the study of CMI in general and of cytotoxic T lymphocytes in particular. Parallels from studies in human and mouse immunology are drawn in order to point out implications to bovine immunology, specifically for immunity against bovine herpesvirus 1.
Collapse
Affiliation(s)
- N R Hegde
- Department of Molecular Microbiology and Immunology, Oregon Health Sciences University, Portland 97201-3098, USA
| | | |
Collapse
|
36
|
Bauer M, Wagner H, Lipford GB. HPV type 16 protein E7 HLA-A2 binding peptides are immunogenic but not processed and presented. Immunol Lett 2000; 71:55-9. [PMID: 10709786 DOI: 10.1016/s0165-2478(99)00170-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Human papillomaviruses (HPV) have been implicated in the etiology of cervical malignancies and a high percentage of cervical carcinoma cells express HPV-16 E6 and E7 oncoproteins. These proteins are attractive targets for cytolytic T lymphocyte (CTL) mediated immunotherapy. We screened peptides derived from the HPV-16 E7 protein for binding to HLA-A2 and tested their potential to induce specific CTL responses in chimeric HLA-A2/H2-Kb transgenic mice. From eight potential binding peptides four displayed binding and were tested for immunogenicity. CTL activity was tested using target cells pulsed with peptide or expressing E7 protein. While there was no CTL induction observed with the peptides 7-15, 66-74 and 82-90, CTL from mice immunized with 86-93 lysed targets presenting the peptide in the context of the HLA-A2/H2-Kb molecule or wild-type HLA-A2. In contrast, 86-93 induced CTL showed no cytolytic activity against cells expressing the protein E7 and vaccination with the E7 protein did not lead to cytotoxicity against targets pulsed with the 86-93 peptide. Therefore the peptide 86-93, which binds to HLA-A2, is able to induce CTL responses in context of HLA-A2, but the peptide appears not to be processed or presented by HPV type 16 infected cells.
Collapse
Affiliation(s)
- M Bauer
- Institute of Medical Microbiology, Immunology and Hygiene, Munich, Germany
| | | | | |
Collapse
|
37
|
Ma H, Ke Y, Li Q, Kapp JA. Bovine and human insulin activate CD8+-autoreactive CTL expressing both type 1 and type 2 cytokines in C57BL/6 mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:86-92. [PMID: 10604997 DOI: 10.4049/jimmunol.164.1.86] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD8+ T cells down-regulate a variety of immune responses. For example, porcine and human insulin do not stimulate Abs in C57BL/6 mice because CD8+ T cells inhibit CD4+ helper T cells. By contrast, bovine insulin induces Ab in C57BL/6 mice, and removal of CD8+ T cells does not alter this response. This raises the question of whether porcine, but not bovine, insulin activates CD8+ T cells or whether both insulins activate CD8+ T cells but CD4+ helper T cells are differentially inhibited by them. In this study, we show that insulin-specific CD8+ CTL can be cultured from C57BL/6 mice primed with either bovine or human insulin in CFA. Thus, exogenous Ags, besides OVA, induce CD8+ CTL when administered in an adjuvant, suggesting this is a typical response. These CTL are H-2Kb restricted and produce IL-5, IL-10, IFN-gamma, and small amounts of IL-4, which is distinct from IFN-gamma and TNF-alpha that are typically secreted by virus-specific CTL. Moreover, the CTL primed with either bovine or human insulin recognize an A-chain peptide that is identical to the mouse insulin sequence. That foreign proteins, which are closely related to self-proteins, activated autoreactive, CD8+ T cells in vivo is a novel finding. It raises the possibility that self-reactive CTL may be activated by cross-reacting Ags and once activated they might participate in autoimmunity. These results also suggest that down-regulation of insulin-specific responses by autoreactive CD8+ T cells is most likely due to the differential sensitivity of bovine and human insulin-specific CD4+ T cells.
Collapse
Affiliation(s)
- H Ma
- Department of Ophthalmology, Winship Cancer Center, Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
38
|
Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S. Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol 1999; 74:181-273. [PMID: 10605607 DOI: 10.1016/s0065-2776(08)60911-6] [Citation(s) in RCA: 816] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- F M Marincola
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
39
|
Kurts C, Sutherland RM, Davey G, Li M, Lew AM, Blanas E, Carbone FR, Miller JF, Heath WR. CD8 T cell ignorance or tolerance to islet antigens depends on antigen dose. Proc Natl Acad Sci U S A 1999; 96:12703-7. [PMID: 10535986 PMCID: PMC23058 DOI: 10.1073/pnas.96.22.12703] [Citation(s) in RCA: 183] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
There are two major mechanisms reported to prevent the autoreactivity of islet-specific CD8(+) T cells: ignorance and tolerance. When ignorance is operative, naïve autoreactive CD8(+) T cells ignore islet antigens and recirculate without causing damage, unless activated by an external stimulus. In the case of tolerance, CD8(+) T cells are deleted. Which factor(s) contributes to each particular outcome was previously unknown. Here, we demonstrate that the concentration of self antigen determines which mechanism operates. When ovalbumin (OVA) was expressed at a relatively low concentration in the pancreatic islets of transgenic mice, there was no detectable cross-presentation, and the CD8(+) T cell compartment remained ignorant of OVA. In mice expressing higher doses of OVA, cross-presentation was detectable and led to peripheral deletion of OVA-specific CD8(+) T cells. When cross-presentation was prevented by reconstituting the bone marrow compartment with cells incapable of presenting OVA, deletional tolerance was converted to ignorance. Thus, the immune system uses two strategies to avoid CD8(+) T cell-mediated autoimmunity: for high dose antigens, it deletes autoreactive T cells, whereas for lower dose antigens, it relies on ignorance.
Collapse
Affiliation(s)
- C Kurts
- Department of Nephrology, Medizinische Hochschule, Carl Neuberg Strasse 1, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Byers DE, Lindahl KF. Peptide Affinity and Concentration Affect the Sensitivity of M3-Restricted CTLs Induced In Vitro. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.6.3022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
In vitro stimulation of mouse splenocytes with hemagglutinin (HA) 173–190, a peptide derived from influenza virus hemagglutinin (A/JAP/305/57, H2N2), induces CTLs that are directed to the MHC class Ib molecule, H2-M3. M3 preferably binds peptides bearing an N-terminal formylmethionine. In this study, we show that several related nonformylated peptides can induce anti-HA CTLs in vitro: MLIIW (the minimal epitope), derived from HA186–190 at the C-terminal end of HA173–190; MLIIWG; MLIIWGV; and MLIIWGI, as well as formylated MLIIW. The heptamer peptides correspond to a polymorphism of HA192 in H2 strains of influenza; they have the highest relative affinities for M3 of the nonformylated peptides and higher affinities than some formylated mitochondrial peptides. Depending on the affinity of the peptide, a range of concentrations can be used to induce CTLs. One nanomolar of the high affinity f-MLIIW peptide can induce anti-HA CTLs, whereas 100-fold more of the lower affinity MLIIW peptide is needed. Lines induced with high concentrations (1 μM or greater) of f-MLIIW recognize Ag poorly, and the most efficient CTLs are induced with the lowest concentrations of peptide. Analysis with a panel of anti-TCRVβ Abs shows that different T cells respond to high vs low peptide; the repertoire of cells responding to higher concentrations is more diverse, consistent with the expansion of more, but less efficient, clones. Thus, peptide affinity and concentration should be considered together for generating efficient antipeptide CTLs in vitro.
Collapse
Affiliation(s)
- Derek E. Byers
- Howard Hughes Medical Institute, Center for Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75235
| | - Kirsten Fischer Lindahl
- Howard Hughes Medical Institute, Center for Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75235
| |
Collapse
|
41
|
Ennis FA, Cruz J, Jameson J, Klein M, Burt D, Thipphawong J. Augmentation of human influenza A virus-specific cytotoxic T lymphocyte memory by influenza vaccine and adjuvanted carriers (ISCOMS). Virology 1999; 259:256-61. [PMID: 10388649 DOI: 10.1006/viro.1999.9765] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is a need to improve the ability of subunit vaccines to induce CD8(+) CTL responses in humans, especially for vaccines used to prevent illness by organisms that undergo antigenic variation at their major neutralizing antibody sites, e.g., influenza A viruses and human immunodeficiency virus. Murine models have demonstrated the protective role of cross-reactive CTL against influenza A virus antigenic drift. We tested the ability of an adjuvanted carrier (Iscomatrix) to help human antigen-presenting cells present formalin-killed influenza vaccine to human CD8(+) CTL clones in vitro and in vaccinated humans. The results of a randomized, double-blind, controlled clinical study demonstrate that a single dose of a vaccine formulated into Iscom particles increased influenza A virus-specific CTL memory in 50-60% of recipients, compared to 5% of the recipients of the standard influenza vaccine.
Collapse
Affiliation(s)
- F A Ennis
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts, 01655, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Santin AD, Hermonat PL, Ravaggi A, Chiriva-Internati M, Zhan D, Pecorelli S, Parham GP, Cannon MJ. Induction of human papillomavirus-specific CD4(+) and CD8(+) lymphocytes by E7-pulsed autologous dendritic cells in patients with human papillomavirus type 16- and 18-positive cervical cancer. J Virol 1999; 73:5402-10. [PMID: 10364287 PMCID: PMC112596 DOI: 10.1128/jvi.73.7.5402-5410.1999] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/1998] [Accepted: 03/24/1999] [Indexed: 02/02/2023] Open
Abstract
Human papillomavirus (HPV) type 16 (HPV 16) and HPV type 18 (HPV 18) are implicated in the induction and progression of the majority of cervical cancers. Since the E6 and E7 oncoproteins of these viruses are expressed in these lesions, such proteins might be potential tumor-specific targets for immunotherapy. In this report, we demonstrate that recombinant, full-length E7-pulsed autologous dendritic cells (DC) can elicit a specific CD8(+) cytotoxic T-lymphocyte (CTL) response against autologous tumor target cells in three patients with HPV 16- or HPV 18-positive cervical cancer. E7-specific CTL populations expressed strong cytolytic activity against autologous tumor cells, did not lyse autologous concanavalin A-treated lymphoblasts or autologous Epstein-Barr virus-transformed lymphoblastoid cell lines (LCL), and showed low levels of cytotoxicity against natural killer cell-sensitive K562 cells. Cytotoxicity against autologous tumor cells could be significantly blocked by anti-HLA class I (W6/32) and anti-CD11a/LFA-1 antibodies. Phenotypically, all CTL populations were CD3(+)/CD8(+), with variable levels of CD56 expression. CTL induced by E7-pulsed DC were also highly cytotoxic against an allogeneic HLA-A2(+) HPV 16-positive matched cell line (CaSki). In addition, we show that specific lymphoproliferative responses by autologous CD4(+) T cells can also be induced by E7-pulsed autologus DC. E7-specific CD4(+) T cells proliferated in response to E7-pulsed LCL but not unpulsed LCL, and this response could be blocked by anti-HLA class II antibody. Finally, with two-color flow cytometric analysis of intracellular cytokine expression at the single-cell level, a marked Th1-like bias (as determined by the frequency of gamma interferon- and interleukin 4-expressing cells) was observable for both CD8(+) and CD4(+) E7-specific lymphocyte populations. Taken together, these data demonstrate that full-length E7-pulsed DC can induce both E7-specific CD4(+) T-cell proliferative responses and strong CD8(+) CTL responses capable of lysing autologous naturally HPV-infected cancer cells in patients with cervical cancer. These results may have important implications for the treatment of cervical cancer patients with active or adoptive immunotherapy.
Collapse
Affiliation(s)
- A D Santin
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Arkansas, Little Rock, Arkansas, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Albert ML, Pearce SF, Francisco LM, Sauter B, Roy P, Silverstein RL, Bhardwaj N. Immature dendritic cells phagocytose apoptotic cells via alphavbeta5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. J Exp Med 1998; 188:1359-68. [PMID: 9763615 PMCID: PMC2212488 DOI: 10.1084/jem.188.7.1359] [Citation(s) in RCA: 936] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/1998] [Revised: 08/11/1998] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells, but not macrophages, efficiently phagocytose apoptotic cells and cross-present viral, tumor, and self-antigens to CD8(+) T cells. This in vitro pathway corresponds to the in vivo phenomena of cross-priming and cross-tolerance. Here, we demonstrate that phagocytosis of apoptotic cells is restricted to the immature stage of dendritic cell (DC) development, and that this process is accompanied by the expression of a unique profile of receptors, in particular the alphavbeta5 integrin and CD36. Upon maturation, these receptors and, in turn, the phagocytic capacity of DCs, are downmodulated. Macrophages engulf apoptotic cells more efficiently than DCs, and although they express many receptors that mediate this uptake, they lack the alphavbeta5 integrin. Furthermore, in contrast to DCs, macrophages fail to cross-present antigenic material contained within the engulfed apoptotic cells. Thus, DCs use unique pathways for the phagocytosis, processing, and presentation of antigen derived from apoptotic cells on class I major histocompatibility complex. We suggest that the alphavbeta5 integrin plays a critical role in the trafficking of exogenous antigen by immature DCs in this cross-priming pathway.
Collapse
Affiliation(s)
- M L Albert
- Laboratory of Cellular Physiology and Immunology, The Rockefeller University, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Wang C, Splitter GA. CD4(+) cytotoxic T-lymphocyte activity against macrophages pulsed with bovine herpesvirus 1 polypeptides. J Virol 1998; 72:7040-7. [PMID: 9696796 PMCID: PMC109924 DOI: 10.1128/jvi.72.9.7040-7047.1998] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bovine herpesvirus 1 (BHV-1) induces immune suppression, but the mechanisms for suppression are not well identified. We examined the induction and activity of BHV-1-specific cytolytic CD4(+) T lymphocytes (CTL) by stimulating peripheral blood mononuclear cells (PBMC) of cattle immunized with attenuated live BHV-1. Cytolytic effector cells were primarily CD4(+) T lymphocytes and lysed autologous, but not allogeneic, macrophages infected with BHV-1 or pulsed with BHV-1 polypeptides. Apoptosis of BHV-1-expressing target cells was observed in CD4(+) CTL assays by terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL) analysis. To determine if apoptosis was mediated by a perforin- or Fas-mediated pathway, EGTA, a known selective inhibitor of the perforin pathway, was used. EGTA did not inhibit CD4(+)-T-cell-mediated cytotoxic activity, but it did limit the NK cell cytotoxicity of virus infected cells. These findings support the concept that CD4(+) CTL lyse macrophages pulsed with BHV-1 polypeptides through a Fas-mediated lytic pathway by inducing apoptosis in the target cells. The prominent cytotoxicity mediated by CD4(+) CTL suggests a mechanism of selective removal of viral antigen-associated antigen-presenting cells.
Collapse
Affiliation(s)
- C Wang
- Department of Animal Health and Biomedical Science, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
45
|
Deng Y, Gibbs J, Bačík I, Porgador A, Copeman J, Lehner P, Ortmann B, Cresswell P, Bennink JR, Yewdell JW. Assembly of MHC Class I Molecules with Biosynthesized Endoplasmic Reticulum-Targeted Peptides Is Inefficient in Insect Cells and Can Be Enhanced by Protease Inhibitors. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.4.1677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
To study the requirements for assembly of MHC class I molecules with antigenic peptides in the endoplasmic reticulum (ER), we studied Ag processing in insect cells. Insects lack a class I recognition system, and their cells therefore provide a “blank slate” for identifying the proteins that have evolved to facilitate assembly of class I molecules in vertebrate cells. H-2Kb heavy chain, mouse β2-microglobulin, and an ER-targeted version of a peptide corresponding to Ova257–264 were expressed in insect cells using recombinant vaccinia viruses. Cell surface expression of Kb-OVA257–264 complexes was quantitated using a recently described complex-specific mAb (25-D1.16). Relative to TAP-deficient human cells, insect cells expressed comparable levels of native, peptide-receptive cell surface Kb molecules, but generated cell surface Kb-OVA257–264 complexes at least 20-fold less efficiently from ER-targeted peptides. The inefficient assembly of Kb-OVA257–264 complexes in the ER of insect cells cannot be attributed solely to a requirement for human tapasin, since first, human cells lacking tapasin expressed endogenously synthesized Kb-OVA257–264 complexes at levels comparable to tapasin-expressing cells, and second, vaccinia virus-mediated expression of human tapasin in insect cells did not detectably enhance the expression of Kb-OVA257–264 complexes. The assembly of Kb-OVA257–264 complexes could be greatly enhanced in insect but not human cells by a nonproteasomal protease inhibitor. These findings indicate that insect cells lack one or more factors required for the efficient assembly of class I-peptide complexes in vertebrate cells and are consistent with the idea that the missing component acts to protect antigenic peptides or their immediate precursors from degradation.
Collapse
Affiliation(s)
| | | | | | - Angel Porgador
- †Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
- Laboratories of
| | - James Copeman
- ‡Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510
- Laboratories of
| | - Paul Lehner
- ‡Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510
- Laboratories of
| | - Bodo Ortmann
- ‡Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510
- Laboratories of
| | - Peter Cresswell
- ‡Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510
- Laboratories of
| | | | | |
Collapse
|
46
|
Obst R, Münz C, Stevanović S, Rammensee HG. Allo- and self-restricted cytotoxic T lymphocytes against a peptide library: evidence for a functionally diverse allorestricted T cell repertoire. Eur J Immunol 1998; 28:2432-43. [PMID: 9710221 DOI: 10.1002/(sici)1521-4141(199808)28:08<2432::aid-immu2432>3.0.co;2-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BALB/c-derived spleen cells were depleted of cytotoxic T lymphocytes (CTL) recognizing allogeneic (H2b) and TAP-negative cells followed by stimulation with the same cells loaded with a synthetic library binding to H2-Kb. The resulting CTL lines were found to differ widely in peptide specificity and to exhibit an avidity towards the library as that demonstrated for syngeneic CTL. These results demonstrate that positive selection in the context of a certain MHC molecule does not seem to be required for generating high-avidity TCR that are restricted by the same molecule. However, positive selection increases the frequency of such CTL. By raising T cell lines from a repertoire which did not undergo negative selection by the restriction element in question, it becomes possible to produce effective self-peptide/ MHC as well as nonself-peptide/MHC-specific CTL as tools for adoptive tumor immunotherapy.
Collapse
Affiliation(s)
- R Obst
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Germany
| | | | | | | |
Collapse
|
47
|
Knight SC, Iqball S, Roberts MS, Macatonia S, Bedford PA. Transfer of antigen between dendritic cells in the stimulation of primary T cell proliferation. Eur J Immunol 1998; 28:1636-44. [PMID: 9603470 DOI: 10.1002/(sici)1521-4141(199805)28:05<1636::aid-immu1636>3.0.co;2-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Primary proliferative T cell responses require stimulation with antigen-pulsed dendritic cells (Ag-DC). Here we show that for optimal stimulation, dendritic cells (DC) not exposed directly to antigen are also required. Ag-DC added to DC-depleted T cells caused negligible primary stimulation; adding back DC resulted in stimulation. These effects were seen using the contact sensitizer fluorescein isothiocyanate (FITC), FITC conjugated to ovalbumin (FITC-OVA) or influenza virus as antigens. DC co-cultured with Ag-DC (using FITC or FITC-OVA) acquired antigen indicating that antigen was transferred between DC. DC that acquired antigen secondarily were separated by cell sorting and stimulated primary T cell proliferation directly. DC were also pulsed with FITC, washed thoroughly and incubated overnight. Supernatants contained shed antigen since DC incubated in these supernatants acquired antigen as indicated by flow cytometry. DC acquiring the shed antigen also stimulated T cell proliferation although the stimulation was not as effective as that seen when cell contact between DC and antigen-bearing DC occurred. Thus, in primary stimulation, activation of T cells may occur when there is an antigen gradient between Ag-DC and DC and the mechanisms underlying these effects are now being sought. We propose that this unique interaction between antigen-presenting cells may be a paradigm for self/non-self discrimination.
Collapse
Affiliation(s)
- S C Knight
- Imperial College School of Medicine, Northwick Park Institute for Medical Research, Harrow, GB.
| | | | | | | | | |
Collapse
|
48
|
Dozmorov IM, Miller RA. Generation of Antigen-Specific Th2 Cells from Unprimed Mice In Vitro: Effects of Dexamethasone and Anti-IL-10 Antibody. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.6.2700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
We describe a system for the in vitro production of Ag-specific mouse CD4 cell lines from unprimed mice. Purified CD4+CD45RBhigh T cells were exposed to Ag-pulsed accessory cells in serum-free medium for 24 h; cultured in the absence of Ag and in the presence of serum, IL-2, dexamethasone, and Abs to IL-10 for an additional 4 days; and then re-exposed to the original sensitizing Ag. The presence of dexamethasone and Abs to IL-10 during the initial expansion stage appeared to be critical for the ability of the stimulated and expanded T cells to respond to restimulation with the same Ag. Repeated cycles of in vitro stimulation led to increased specificity for the sensitizing Ag (in the current case, pigeon cytochrome c), a decline in production of IL-2 and IFN-γ, and increased production of IL-4, IL-5, and IL-10. This culture protocol provides a test system for exploration of factors that regulate the conversion of naïve cells to memory cells and the development of specific immune responses to protein Ags. The data are consistent with models that implicate glucocorticoids as regulators of immune response specificity.
Collapse
Affiliation(s)
| | - Richard A. Miller
- *Department of Pathology,
- †Geriatrics Center, and
- ‡Institute of Gerontology, University of Michigan, Ann Arbor, MI 48109; and
- §Ann Arbor DVA Medical Center, Ann Arbor, MI 48109
| |
Collapse
|
49
|
White DW, Harty JT. Perforin-Deficient CD8+ T Cells Provide Immunity to Listeria monocytogenes by a Mechanism That Is Independent of CD95 and IFN-γ but Requires TNF-α. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.2.898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
CD8+ T cells are effective mediators of immunity against Listeria monocytogenes, but the mechanisms by which they provide antilisterial immunity are poorly understood. CD8+ T cells efficiently lyse target cells in vitro by at least two independent pathways. To test the hypothesis that CD8+ T cell-mediated immunity to L. monocytogenes is dependent on perforin or CD95 (Fas, Apo-1), we used C57Bl/6 (B6) and perforin-deficient (PO) mice to generate CD8+ T cell lines specific for the L. monocytogenes-encoded Ag listeriolysin O (LLO). Both lines specifically produce IFN-γ and TNF-α, and mediate target cell lysis in vitro. Cytolysis mediated by the PO-derived CD8+ T cell line is delayed relative to the B6-derived line and is completely inhibited by anti-CD95 Abs. In vivo, PO-derived CD8+ T cells provide specific antilisterial immunity in B6 hosts, CD95-deficient hosts, and IFN-γ-depleted hosts. However, PO-derived CD8+ T cells fail to provide antilisterial immunity in hosts depleted of TNF-α. These results indicate that single Ag-specific CD8+ T cells derived from PO mice can mediate antilisterial immunity by a mechanism that is independent of CD95 or IFN-γ, but requires TNF-α.
Collapse
Affiliation(s)
| | - John T. Harty
- *Interdisciplinary Graduate Program in Immunology and
- †Department of Microbiology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
50
|
Speidel K, Osen W, Faath S, Hilgert I, Obst R, Braspenning J, Momburg F, Hämmerling GJ, Rammensee HG. Priming of cytotoxic T lymphocytes by five heat-aggregated antigens in vivo: conditions, efficiency, and relation to antibody responses. Eur J Immunol 1997; 27:2391-9. [PMID: 9341785 DOI: 10.1002/eji.1830270938] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Mice were immunized i.p. with soluble or heat-denatured protein antigens [ovalbumin, beta-galactosidase, or recombinant E7 protein of human papilloma virus type 16 (HBV)]. Heat-denatured (100 degrees C) preparations of these proteins were able to induce cytotoxic T lymphocytes (CTL) that recognize cells expressing the respective genes, whereas native protein was either inefficient or required up to 30-fold higher doses. If the heat-treated proteins were separated into aggregated and soluble fractions by ultracentrifugation, only the aggregated fractions were able to induce specific CTL; this is probably because of the easier access to one of the major histocompatibility complex class I loading pathways for exogenous antigen. Addition of the adjuvant aluminium hydroxide (alum) to aggregated proteins abolished their ability to induce CTL; thus, a condition leading to a strong antibody response appeared to inhibit CTL induction. Interestingly, immunization with heat-denatured ovalbumin plus alum increased the IgM/IgG1 ratio compared to immunization with native ovalbumin and alum. Immunization of B6 mice transgenic for an HLA-A2/H-2K(b) hybrid gene with heat-denatured, recombinant HPV 16-E7 protein induced D(b)-restricted CTL specific for the peptide 49-57 of E7, indicating that this epitope is immunodominant over any A2-restricted E7 epitope in these mice. A whole influenza virus preparation heated to 100 degrees C or even autoclaved was still able to induce virus-specific CTL and BALB/c spleen cells heated to 100 degrees C could still cross-prime minor H-specific CTL in B6 mice, although with lower efficiency than fresh spleen cells. Thus, aggregated proteins can be considered as components for future vaccines.
Collapse
Affiliation(s)
- K Speidel
- Department of Tumorvirus Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|