1
|
Vyasamneni R, Kohler V, Karki B, Mahimkar G, Esaulova E, McGee J, Kallin D, Sheen JH, Harjanto D, Kirsch M, Poran A, Dong J, Srinivasan L, Gaynor RB, Bushway ME, Srouji JR. A universal MHCII technology platform to characterize antigen-specific CD4 + T cells. CELL REPORTS METHODS 2023; 3:100388. [PMID: 36814840 PMCID: PMC9939426 DOI: 10.1016/j.crmeth.2022.100388] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/08/2022] [Accepted: 12/20/2022] [Indexed: 01/15/2023]
Abstract
CD4+ T cells are critical to the immune system and perform multiple functions; therefore, their identification and characterization are crucial to better understanding the immune system in both health and disease states. However, current methods rarely preserve their ex vivo phenotype, thus limiting our understanding of their in vivo functions. Here we introduce a flexible, rapid, and robust platform for ex vivo CD4+ T cell identification. By combining MHCII allele purification, allele-independent peptide loading, and multiplexed flow cytometry technologies, we can enable high-throughput personalized CD4+ T cell identification, immunophenotyping, and sorting. Using this platform in combination with single-cell sorting and multimodal analyses, we identified and characterized antigen-specific CD4+ T cells relevant to COVID-19 and cancer neoantigen immunotherapy. Overall, our platform can be used to detect and characterize CD4+ T cells across multiple diseases, with potential to guide CD4+ T cell epitope design for any disease-specific immunization strategy.
Collapse
Affiliation(s)
| | | | - Binisha Karki
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | - Gauri Mahimkar
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | | | - Jonathan McGee
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | - Daniel Kallin
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | | | - Dewi Harjanto
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | - Miles Kirsch
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | - Asaf Poran
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | - Jesse Dong
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| | | | | | | | - John R. Srouji
- BioNTech US, Inc., 40 Erie Street, Cambridge, MA 02139, USA
| |
Collapse
|
2
|
Luo G, Yogeshwar S, Lin L, Mignot EJM. T cell reactivity to regulatory factor X4 in type 1 narcolepsy. Sci Rep 2021; 11:7841. [PMID: 33837283 PMCID: PMC8035403 DOI: 10.1038/s41598-021-87481-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/23/2021] [Indexed: 11/11/2022] Open
Abstract
Type 1 narcolepsy is strongly (98%) associated with human leukocyte antigen (HLA) class II DQA1*01:02/DQB1*06:02 (DQ0602) and highly associated with T cell receptor (TCR) alpha locus polymorphism as well as other immune regulatory loci. Increased incidence of narcolepsy was detected following the 2009 H1N1 pandemic and linked to Pandemrix vaccination, strongly supporting that narcolepsy is an autoimmune disorder. Although recent results suggest CD4+ T cell reactivity to neuropeptide hypocretin/orexin and cross-reactive flu peptide is involved, identification of other autoantigens has remained elusive. Here we study whether autoimmunity directed against Regulatory Factor X4 (RFX4), a protein co-localized with hypocretin, is involved in some cases of narcolepsy. Studying human serum, we found that autoantibodies against RFX4 were rare. Using RFX4 peptides bound to DQ0602 tetramers, antigen RFX4-86, -95, and -60 specific human CD4+ T cells were detected in 4/10 patients and 2 unaffected siblings, but not in others. Following culture with each cognate peptide, enriched autoreactive TCRαβ clones were isolated by single-cell sorting and TCR sequenced. Homologous clones bearing TRBV4-2 and recognizing RFX4-86 in patients and one twin control of patient were identified. These results suggest the involvement of RFX4 CD4+ T cell autoreactivity in some cases of narcolepsy, but also in healthy donors.
Collapse
Affiliation(s)
- Guo Luo
- Department of Psychiatry and Behavioral Sciences, Stanford University Center for Sleep Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Selina Yogeshwar
- Department of Psychiatry and Behavioral Sciences, Stanford University Center for Sleep Sciences, Stanford University School of Medicine, Palo Alto, CA, USA.,Division of Biosciences, Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ling Lin
- Department of Psychiatry and Behavioral Sciences, Stanford University Center for Sleep Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Emmanuel Jean-Marie Mignot
- Department of Psychiatry and Behavioral Sciences, Stanford University Center for Sleep Sciences, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
3
|
Archila LD, Chow IT, McGinty JW, Renand A, Jeong D, Robinson D, Farrington ML, Kwok WW. Ana o 1 and Ana o 2 cashew allergens share cross-reactive CD4(+) T cell epitopes with other tree nuts. Clin Exp Allergy 2017; 46:871-83. [PMID: 27129138 DOI: 10.1111/cea.12746] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Allergies to cashew are increasing in prevalence, with clinical symptoms ranging from oral pruritus to fatal anaphylactic reaction. Yet, cashew-specific T cell epitopes and T cell cross-reactivity amongst cashew and other tree nut allergens in humans remain uncharacterized. OBJECTIVES In this study, we characterized cashew-specific T cell responses in cashew-allergic subjects and examined cross-reactivity of these cashew-specific cells towards other tree nut allergens. METHODS CD154 up-regulation assay was used to determine immunodominance hierarchy among cashew major allergens at the T cell level. The phenotype, magnitude and functionality of cashew-specific T cells were determined by utilizing ex vivo staining with MHC class II tetramers. Dual tetramer staining and proliferation experiments were used to determine cross-reactivity to other tree nuts. RESULTS CD4(+) T cell responses were directed towards cashew allergens Ana o 1 and Ana o 2. Multiple Ana o 1 and Ana o 2 T cell epitopes were then identified. These epitopes elicited either TH 2 or TH 2/TH 17 responses in allergic subjects, which were either cashew unique epitope or cross-reactive epitopes. For clones that recognized the cross-reactive epitope, T cell clones responded robustly to cashew, hazelnut and/or pistachio but not to walnut. CONCLUSIONS Phylogenetically diverse tree nut allergens can activate cashew-reactive T cells and elicit a TH 2-type response at an epitope-specific level. CLINICAL RELEVANCE Lack of cross-reactivity between walnut and cashew suggests that cashew peptide immunotherapy approach may not be most effective for walnut.
Collapse
Affiliation(s)
- L D Archila
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - I-T Chow
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - J W McGinty
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - A Renand
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - D Jeong
- Virginia Mason Medical center, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - D Robinson
- Virginia Mason Medical center, Seattle, WA, USA
| | | | - W W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
4
|
Spanier JA, Sahli NL, Wilson JC, Martinov T, Dileepan T, Burrack AL, Finger EB, Blazar BR, Michels AW, Moran A, Jenkins MK, Fife BT. Increased Effector Memory Insulin-Specific CD4 + T Cells Correlate With Insulin Autoantibodies in Patients With Recent-Onset Type 1 Diabetes. Diabetes 2017; 66:3051-3060. [PMID: 28842400 PMCID: PMC5697953 DOI: 10.2337/db17-0666] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/18/2017] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) results from T cell-mediated destruction of insulin-producing β-cells. Insulin represents a key self-antigen in disease pathogenesis, as recent studies identified proinsulin-responding T cells from inflamed pancreatic islets of organ donors with recent-onset T1D. These cells respond to an insulin B-chain (InsB) epitope presented by the HLA-DQ8 molecule associated with high T1D risk. Understanding insulin-specific T-cell frequency and phenotype in peripheral blood is now critical. We constructed fluorescent InsB10-23:DQ8 tetramers, stained peripheral blood lymphocytes directly ex vivo, and show DQ8+ patients with T1D have increased tetramer+ CD4+ T cells compared with HLA-matched control subjects without diabetes. Patients with a shorter disease duration had higher frequencies of insulin-reactive CD4+ T cells, with most of these cells being antigen experienced. We also demonstrate that the number of insulin tetramer+ effector memory cells is directly correlated with insulin antibody titers, suggesting insulin-specific T- and B-cell interactions. Notably, one of four control subjects with tetramer+ cells was a first-degree relative who had insulin-specific cells with an effector memory phenotype, potentially representing an early marker of T-cell autoimmunity. Our results suggest that studying InsB10-23:DQ8 reactive T-cell frequency and phenotype may provide a biomarker of disease activity in patients with T1D and those at risk.
Collapse
Affiliation(s)
- Justin A Spanier
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Nathanael L Sahli
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Joseph C Wilson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Thamotharampillai Dileepan
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Adam L Burrack
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Erik B Finger
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN
| | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Aaron W Michels
- Department of Pediatrics and Medicine, University of Colorado, Denver, CO
| | - Antoinette Moran
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
5
|
Absence of autoreactive CD4 + T-cells targeting HLA-DQA1*01:02/DQB1*06:02 restricted hypocretin/orexin epitopes in narcolepsy type 1 when detected by EliSpot. J Neuroimmunol 2017; 309:7-11. [DOI: 10.1016/j.jneuroim.2017.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 11/20/2022]
|
6
|
Archila LD, Khan FS, Bhatnagar N, Robinson D, Farrington ML, Kwok WW. α S1-Casein elucidate major T-cell responses in cow's milk allergy. J Allergy Clin Immunol 2017; 140:854-857.e6. [PMID: 28343843 DOI: 10.1016/j.jaci.2017.02.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/30/2017] [Accepted: 02/07/2017] [Indexed: 02/06/2023]
Affiliation(s)
- L Diego Archila
- Benaroya Research Institute at Virginia Mason, Seattle, Wash
| | - Fatima S Khan
- Benaroya Research Institute at Virginia Mason, Seattle, Wash; Department of Medicine, University of Washington, Seattle, Wash
| | - Nupur Bhatnagar
- Benaroya Research Institute at Virginia Mason, Seattle, Wash
| | | | | | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, Wash; Department of Medicine, University of Washington, Seattle, Wash.
| |
Collapse
|
7
|
Mazor R, Addissie S, Jang Y, Tai CH, Rose J, Hakim F, Pastan I. Role of HLA-DP in the Presentation of Epitopes from the Truncated Bacterial PE38 Immunotoxin. AAPS JOURNAL 2016; 19:117-129. [PMID: 27796910 DOI: 10.1208/s12248-016-9986-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023]
Abstract
Identification of helper T-cell epitopes is important in many fields of medicine. We previously used an experimental approach to identify T-cell epitopes in PE38, a truncated bacterial toxin used in immunotoxins. Here, we evaluated the ability of antibodies to DR, DP, or DQ to block T-cell responses to PE38 epitopes in 36 PBMC samples. We predicted the binding affinities of peptides to DR, DP, and DQ alleles using computational tools and analyzed their ability to predict the T-cell epitopes. We found that HLA-DR is responsible for 65% of the responses, DP 24%, and DQ 4%. One epitope that is presented in 20% of the samples (10/50) is entirely DP restricted and was not predicted to bind to DR or DP reference alleles using binding algorithms. We conclude that DP has an important role in helper T-cell response to PE38.
Collapse
Affiliation(s)
- Ronit Mazor
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5106, Bethesda, Maryland, 20892-4264, USA
| | - Selamawit Addissie
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5106, Bethesda, Maryland, 20892-4264, USA
| | - Youjin Jang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5106, Bethesda, Maryland, 20892-4264, USA
| | - Chin-Hsien Tai
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5106, Bethesda, Maryland, 20892-4264, USA
| | - Jeremy Rose
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Fran Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Room 5106, Bethesda, Maryland, 20892-4264, USA.
| |
Collapse
|
8
|
Jing L, Laing KJ, Dong L, Russell RM, Barlow RS, Haas JG, Ramchandani MS, Johnston C, Buus S, Redwood AJ, White KD, Mallal SA, Phillips EJ, Posavad CM, Wald A, Koelle DM. Extensive CD4 and CD8 T Cell Cross-Reactivity between Alphaherpesviruses. THE JOURNAL OF IMMUNOLOGY 2016; 196:2205-2218. [PMID: 26810224 DOI: 10.4049/jimmunol.1502366] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/15/2015] [Indexed: 12/18/2022]
Abstract
The Alphaherpesvirinae subfamily includes HSV types 1 and 2 and the sequence-divergent pathogen varicella zoster virus (VZV). T cells, controlled by TCR and HLA molecules that tolerate limited epitope amino acid variation, might cross-react between these microbes. We show that memory PBMC expansion with either HSV or VZV enriches for CD4 T cell lines that recognize the other agent at the whole-virus, protein, and peptide levels, consistent with bidirectional cross-reactivity. HSV-specific CD4 T cells recovered from HSV-seronegative persons can be explained, in part, by such VZV cross-reactivity. HSV-1-reactive CD8 T cells also cross-react with VZV-infected cells, full-length VZV proteins, and VZV peptides, as well as kill VZV-infected dermal fibroblasts. Mono- and cross-reactive CD8 T cells use distinct TCRB CDR3 sequences. Cross-reactivity to VZV is reconstituted by cloning and expressing TCRA/TCRB receptors from T cells that are initially isolated using HSV reagents. Overall, we define 13 novel CD4 and CD8 HSV-VZV cross-reactive epitopes and strongly imply additional cross-reactive peptide sets. Viral proteins can harbor both CD4 and CD8 HSV/VZV cross-reactive epitopes. Quantitative estimates of HSV/VZV cross-reactivity for both CD4 and CD8 T cells vary from 10 to 50%. Based on these findings, we hypothesize that host herpesvirus immune history may influence the pathogenesis and clinical outcome of subsequent infections or vaccinations for related pathogens and that cross-reactive epitopes and TCRs may be useful for multi-alphaherpesvirus vaccine design and adoptive cellular therapy.
Collapse
Affiliation(s)
- Lichen Jing
- Department of Medicine, University of Washington, Seattle, USA
| | - Kerry J Laing
- Department of Medicine, University of Washington, Seattle, USA
| | - Lichun Dong
- Department of Medicine, University of Washington, Seattle, USA
| | | | - Russell S Barlow
- Department of Global Health, University of Washington, Seattle, USA
| | - Juergen G Haas
- Max von Pettenkofer-Institute, Munich, Germany.,Division of Pathway Medicine, University of Edinburgh, United Kingdom
| | | | | | - Soren Buus
- Laboratory of Experimental Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Alec J Redwood
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia
| | - Katie D White
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
| | - Simon A Mallal
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
| | - Elizabeth J Phillips
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, USA
| | - Christine M Posavad
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Department of Laboratory Medicine, University of Washington, Seattle, USA
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle, USA.,Department of Epidemiology, University of Washington, Seattle, USA.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Department of Laboratory Medicine, University of Washington, Seattle, USA
| | - David M Koelle
- Department of Medicine, University of Washington, Seattle, USA.,Department of Global Health, University of Washington, Seattle, USA.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, USA.,Department of Laboratory Medicine, University of Washington, Seattle, USA.,Benaroya Research Institute, Seattle, USA
| |
Collapse
|
9
|
Mannering SI, Pathiraja V, Kay TWH. The case for an autoimmune aetiology of type 1 diabetes. Clin Exp Immunol 2015; 183:8-15. [PMID: 26313217 DOI: 10.1111/cei.12699] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2015] [Indexed: 01/10/2023] Open
Abstract
Type 1 diabetes (T1D) develops when there are insufficient insulin-producing beta cells to maintain glucose homeostasis. The prevailing view has been that T1D is caused by immune-mediated destruction of the pancreatic beta cells. However, several recent papers have challenged the long-standing paradigm describing T1D as a tissue-specific autoimmune disease. These authors have highlighted the gaps in our knowledge and understanding of the aetiology of T1D in humans. Here we review the evidence and argue the case for the autoimmune basis of human T1D. In particular, recent analysis of human islet-infiltrating T cells brings important new evidence to this question. Further data in support of the autoimmune basis of T1D from many fields, including genetics, experimental therapies and immunology, is discussed. Finally, we highlight some of the persistent questions relating to the pathogenesis of human type 1 diabetes that remain to be answered.
Collapse
Affiliation(s)
- S I Mannering
- Immunology and Diabetes Unit, St Vincent's Institute of Medical Research.,Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - V Pathiraja
- Immunology and Diabetes Unit, St Vincent's Institute of Medical Research.,Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - T W H Kay
- Immunology and Diabetes Unit, St Vincent's Institute of Medical Research.,Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| |
Collapse
|
10
|
Nayak K, Jing L, Russell RM, Davies DH, Hermanson G, Molina DM, Liang X, Sherman DR, Kwok WW, Yang J, Kenneth J, Ahamed SF, Chandele A, Murali-Krishna K, Koelle DM. Identification of novel Mycobacterium tuberculosis CD4 T-cell antigens via high throughput proteome screening. Tuberculosis (Edinb) 2015; 95:275-87. [PMID: 25857935 DOI: 10.1016/j.tube.2015.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 02/24/2015] [Accepted: 03/01/2015] [Indexed: 10/23/2022]
Abstract
Elicitation of CD4 IFN-gamma T cell responses to Mycobacterium tuberculosis (MTB) is a rational vaccine strategy to prevent clinical tuberculosis. Diagnosis of MTB infection is based on T-cell immune memory to MTB antigens. The MTB proteome contains over four thousand open reading frames (ORFs). We conducted a pilot antigen identification study using 164 MTB proteins and MTB-specific T-cells expanded in vitro from 12 persons with latent MTB infection. Enrichment of MTB-reactive T-cells from PBMC used cell sorting or an alternate system compatible with limited resources. MTB proteins were used as single antigens or combinatorial matrices in proliferation and cytokine secretion readouts. Overall, our study found that 44 MTB proteins were antigenic, including 27 not previously characterized as CD4 T-cell antigens. Antigen truncation, peptide, NTM homology, and HLA class II tetramer studies confirmed malate synthase G (encoded by gene Rv1837) as a CD4 T-cell antigen. This simple, scalable system has potential utility for the identification of candidate MTB vaccine and biomarker antigens.
Collapse
Affiliation(s)
- Kaustuv Nayak
- ICGEB-Emory Vaccine Center, International Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Lichen Jing
- Department of Medicine, Division of Infectious Diseases, University of Washington, Box 358061, Seattle, WA 98195, USA.
| | - Ronnie M Russell
- Department of Medicine, Division of Infectious Diseases, University of Washington, Box 358061, Seattle, WA 98195, USA.
| | - D Huw Davies
- Department of Medicine, Division of Infectious Diseases, University of California, Room 376D Med-Surg II, Irvine, CA 92697-4068, USA; Antigen Discovery, Inc., 1 Technology Drive Suite E309, Irvine, CA 92618, USA.
| | - Gary Hermanson
- Antigen Discovery, Inc., 1 Technology Drive Suite E309, Irvine, CA 92618, USA.
| | - Douglas M Molina
- Antigen Discovery, Inc., 1 Technology Drive Suite E309, Irvine, CA 92618, USA.
| | - Xiaowu Liang
- Antigen Discovery, Inc., 1 Technology Drive Suite E309, Irvine, CA 92618, USA.
| | - David R Sherman
- Seattle Biomedical Research Institute, 307 Westlake Ave. North, No. 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Box 359931, Seattle, WA 98195, USA.
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA, 98101, USA.
| | - Junbao Yang
- Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA, 98101, USA.
| | - John Kenneth
- Division of Infectious Diseases, St. John's Research Institute, St. John's National Academy of Health Sciences, Sarjapur Road, Koramangala 2 Block, Bangaluru, Karnataka 560034, India.
| | - Syed F Ahamed
- Division of Infectious Diseases, St. John's Research Institute, St. John's National Academy of Health Sciences, Sarjapur Road, Koramangala 2 Block, Bangaluru, Karnataka 560034, India.
| | - Anmol Chandele
- ICGEB-Emory Vaccine Center, International Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India; Emory Vaccine Center, 1510 Clifton Road, Atlanta, GA 30329, USA.
| | - Kaja Murali-Krishna
- ICGEB-Emory Vaccine Center, International Center for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India; Emory Vaccine Center, 1510 Clifton Road, Atlanta, GA 30329, USA; Department of Pediatrics, Emory University, 1760 Haygood Drive, Atlanta, GA 30322, USA.
| | - David M Koelle
- Department of Medicine, Division of Infectious Diseases, University of Washington, Box 358061, Seattle, WA 98195, USA; Department of Global Health, University of Washington, Box 359931, Seattle, WA 98195, USA; Benaroya Research Institute at Virginia Mason, 1201 9th Ave., Seattle, WA, 98101, USA; Department of Laboratory Medicine, University of Washington, Box 358070, Seattle, WA 98195, USA; Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, 1100 Eastlake Ave. East, Seattle, WA 98109, USA.
| |
Collapse
|
11
|
Jug r 2-reactive CD4(+) T cells have a dominant immune role in walnut allergy. J Allergy Clin Immunol 2015; 136:983-92.e7. [PMID: 25772597 PMCID: PMC4568181 DOI: 10.1016/j.jaci.2015.01.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 01/20/2015] [Accepted: 01/22/2015] [Indexed: 12/24/2022]
Abstract
Background Allergic reactions to walnut can be life threatening. While IgE epitopes of walnut have been studied, CD4+ T-cell specific epitopes for walnut remain uncharacterized. Particularly, the relationship of both phenotype and frequency of walnut specific T-cells to the disease have not been examined. Objectives We sought to provide a thorough phenotypic analysis for walnut reactive T-cells in allergic and non-allergic subjects. Particularly, the relationship of phenotypes and frequencies of walnut specific T-cells with the disease. Methods CD154 up-regulation assay was used to examine CD4+ T-cell reactivity towards walnut allergens.Jug r 1, Jug r 2 and Jug r 3. Tetramer-Guided epitope mapping approach was utilized to identify HLA-restricted CD4+ T-cells epitopes in Jug r 2. Direct ex vivo staining with peptide-major histocompatibility complex class II (pMHC-II) tetramers enabled the comparison of frequency and phenotype of Jug r 2-specific CD4+ T-cells between allergic and non-allergic subjects. Jug r 2-specific T-cell-clones were also generated and mRNA transcription factor levels were assessed by RT qPCR. Intracellular cytokine staining (ICS) assays were performed for further phenotypical analyses. Results Jug r 2 was identified as the major allergen that elicited CD4+ T-cell responses. Multiple Jug r 2 T-cell epitopes were identified. The majority of these T-cells in allergic subjects have a CCR4+ TCM (central memory) phenotype. A subset of these T-cells express CCR4+CCR6+ irrespectively of the asthmatic status of the allergic subjects. ICS confirmed these TH2, TH2/TH17 and TH17-like heterogenic profiles. Jug r 2-specific T-cell-clones from allergic subjects mainly expressed GATA3; nonetheless, a portion of T-cell clones expressed either GATA3 and RORC, or RORC, confirming the presence of TH2, TH2/TH17 and TH17 cells. Conclusions Jug r 2 specific responses dominate walnut T-cell responses in subjects with walnut allergy. Jug r 2 central memory CD4+ cells and terminal effector T-cells were detected in peripheral blood with the central memory phenotype as the most prevalent phenotype. In addition to conventional TH2-cells, TH2/TH17 and TH17 cells were also detected in non-asthmatic and asthmatic subjects with walnut allergy. Understanding this T-cell heterogeneity may render better understanding of the disease manifestation.
Collapse
|
12
|
Ascough S, Ingram RJ, Chu KK, Reynolds CJ, Musson JA, Doganay M, Metan G, Ozkul Y, Baillie L, Sriskandan S, Moore SJ, Gallagher TB, Dyson H, Williamson ED, Robinson JH, Maillere B, Boyton RJ, Altmann DM. Anthrax lethal factor as an immune target in humans and transgenic mice and the impact of HLA polymorphism on CD4+ T cell immunity. PLoS Pathog 2014; 10:e1004085. [PMID: 24788397 PMCID: PMC4006929 DOI: 10.1371/journal.ppat.1004085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 03/07/2014] [Indexed: 11/23/2022] Open
Abstract
Bacillus anthracis produces a binary toxin composed of protective antigen (PA) and one of two subunits, lethal factor (LF) or edema factor (EF). Most studies have concentrated on induction of toxin-specific antibodies as the correlate of protective immunity, in contrast to which understanding of cellular immunity to these toxins and its impact on infection is limited. We characterized CD4+ T cell immunity to LF in a panel of humanized HLA-DR and DQ transgenic mice and in naturally exposed patients. As the variation in antigen presentation governed by HLA polymorphism has a major impact on protective immunity to specific epitopes, we examined relative binding affinities of LF peptides to purified HLA class II molecules, identifying those regions likely to be of broad applicability to human immune studies through their ability to bind multiple alleles. Transgenics differing only in their expression of human HLA class II alleles showed a marked hierarchy of immunity to LF. Immunogenicity in HLA transgenics was primarily restricted to epitopes from domains II and IV of LF and promiscuous, dominant epitopes, common to all HLA types, were identified in domain II. The relevance of this model was further demonstrated by the fact that a number of the immunodominant epitopes identified in mice were recognized by T cells from humans previously infected with cutaneous anthrax and from vaccinated individuals. The ability of the identified epitopes to confer protective immunity was demonstrated by lethal anthrax challenge of HLA transgenic mice immunized with a peptide subunit vaccine comprising the immunodominant epitopes that we identified. Anthrax is of concern with respect to human exposure in endemic regions, concerns about bioterrorism and the considerable global burden of livestock infections. The immunology of this disease remains poorly understood. Vaccination has been based on B. anthracis filtrates or attenuated spore-based vaccines, with more recent trials of next-generation recombinant vaccines. Approaches generally require extensive vaccination regimens and there have been concerns about immunogenicity and adverse reactions. An ongoing need remains for rationally designed, effective and safe anthrax vaccines. The importance of T cell stimulating vaccines is inceasingly recognized. An essential step is an understanding of immunodominant epitopes and their relevance across the diverse HLA immune response genes of human populations. We characterized CD4 T cell immunity to anthrax Lethal Factor (LF), using HLA transgenic mice, as well as testing candidate peptide epitopes for binding to a wide range of HLA alleles. We identified anthrax epitopes, noteworthy in that they elicit exceptionally strong immunity with promiscuous binding across multiple HLA alleles and isotypes. T cell responses in humans exposed to LF through either natural anthrax infection or vaccination were also examined. Epitopes identified as candidates were used to protect HLA transgenic mice from anthrax challenge.
Collapse
Affiliation(s)
- Stephanie Ascough
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Rebecca J. Ingram
- Centre for Infection and Immunity, Queen's University Belfast, Belfast, United Kingdom
| | - Karen K. Chu
- Department of Medicine, Imperial College London, London, United Kingdom
| | | | - Julie A. Musson
- Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mehmet Doganay
- Department of Infectious Disease, Erciyes University Hospital, Kayseri, Turkey
| | - Gökhan Metan
- Department of Infectious Disease, Erciyes University Hospital, Kayseri, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, Erciyes University Hospital, Kayseri, Turkey
| | - Les Baillie
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | | | - Stephen J. Moore
- BIOMET, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Theresa B. Gallagher
- BIOMET, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Hugh Dyson
- Defence Science Technology Laboratory, Porton Down, Salisbury, United Kingdom
| | - E. Diane Williamson
- Defence Science Technology Laboratory, Porton Down, Salisbury, United Kingdom
| | - John H. Robinson
- Institute for Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Bernard Maillere
- CEA, iBiTecS, Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), Gif Sur Yvette, France
| | | | - Daniel M. Altmann
- Department of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Virologic and immunologic evidence of multifocal genital herpes simplex virus 2 infection. J Virol 2014; 88:4921-31. [PMID: 24554666 DOI: 10.1128/jvi.03285-13] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Genital herpes simplex virus (HSV) reactivation is thought to be anatomically and temporally localized, coincident with limited ganglionic infection. Short, subclinical shedding episodes are the most common form of HSV-2 reactivation, with host clearance mechanisms leading to rapid containment. The anatomic distribution of shedding episodes has not been characterized. To precisely define patterns of anatomic reactivation, we divided the genital tract into a 22-region grid and obtained daily swabs for 20 days from each region in 28 immunocompetent, HSV-2-seropositive persons. HSV was detected via PCR, and sites of asymptomatic HSV shedding were subjected to a biopsy procedure within 24 h. CD4(+) and CD8(+) T cells were quantified by immunofluorescence, and HSV-specific CD4(+) T cells were identified by intracellular cytokine cytometry. HSV was detected in 868 (7%) of 11,603 genital swabs at a median of 12 sites per person (range, 0 to 22). Bilateral HSV detection occurred on 83 (67%) days with shedding, and the median quantity of virus detected/day was associated with the number of sites positive (P < 0.001). In biopsy specimens of asymptomatic shedding sites, we found increased numbers of CD8(+) T cells compared to control tissue (27 versus 13 cells/mm(2), P = 0.03) and identified HSV-specific CD4(+) T cells. HSV reactivations emanate from widely separated anatomic regions of the genital tract and are associated with a localized cellular infiltrate that was demonstrated to be HSV specific in 3 cases. These data provide evidence that asymptomatic HSV-2 shedding contributes to chronic inflammation throughout the genital tract. IMPORTANCE This detailed report of the anatomic patterns of genital HSV-2 shedding demonstrates that HSV-2 reactivation can be detected at multiple bilateral sites in the genital tract, suggesting that HSV establishes latency throughout the sacral ganglia. In addition, genital biopsy specimens from sites of asymptomatic HSV shedding have increased numbers of CD8(+) T cells compared to control tissue, and HSV-specific CD4(+) T cells are found at sites of asymptomatic shedding. These findings suggest that widespread asymptomatic genital HSV-2 shedding is associated with a targeted host immune response and contributes to chronic inflammation throughout the genital tract.
Collapse
|
14
|
Braendstrup P, Justesen S, Østerbye T, Nielsen LLB, Mallone R, Vindeløv L, Stryhn A, Buus S. MHC class II tetramers made from isolated recombinant α and β chains refolded with affinity-tagged peptides. PLoS One 2013; 8:e73648. [PMID: 24023895 PMCID: PMC3759463 DOI: 10.1371/journal.pone.0073648] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 07/22/2013] [Indexed: 12/24/2022] Open
Abstract
Targeting CD4+ T cells through their unique antigen-specific, MHC class II-restricted T cell receptor makes MHC class II tetramers an attractive strategy to identify, validate and manipulate these cells at the single cell level. Currently, generating class II tetramers is a specialized undertaking effectively limiting their use and emphasizing the need for improved methods of production. Using class II chains expressed individually in E. coli as versatile recombinant reagents, we have previously generated peptide-MHC class II monomers, but failed to generate functional class II tetramers. Adding a monomer purification principle based upon affinity-tagged peptides, we here provide a robust method to produce class II tetramers and demonstrate staining of antigen-specific CD4+ T cells. We also provide evidence that both MHC class II and T cell receptor molecules largely accept affinity-tagged peptides. As a general approach to class II tetramer generation, this method should support rational CD4+ T cell epitope discovery as well as enable specific monitoring and manipulation of CD4+ T cell responses.
Collapse
Affiliation(s)
- Peter Braendstrup
- Laboratory of Experimental Immunology, University of Copenhagen, Copenhagen, Denmark
- Allogeneic Hematopoietic Cell Transplantation Laboratory, Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Sune Justesen
- Laboratory of Experimental Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Østerbye
- Laboratory of Experimental Immunology, University of Copenhagen, Copenhagen, Denmark
| | | | - Roberto Mallone
- Institut National de la Santé et de la Recherche Médicale, Unité 1016, Cochin Institute, DeAR Lab Avenir, Paris, France
| | - Lars Vindeløv
- Allogeneic Hematopoietic Cell Transplantation Laboratory, Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Anette Stryhn
- Laboratory of Experimental Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Søren Buus
- Laboratory of Experimental Immunology, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
15
|
Weder P, Schumacher TNM, Spits H, Luiten RM. Testing for HLA/peptide tetramer-binding to the T cell receptor complex on human T lymphocytes. RESULTS IN IMMUNOLOGY 2012; 2:88-96. [PMID: 24371571 DOI: 10.1016/j.rinim.2012.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 04/27/2012] [Accepted: 04/27/2012] [Indexed: 10/28/2022]
Abstract
HLA/peptide tetramers are frequently used for ex vivo monitoring of disease- or vaccine-induced T cell immune responses and for T cell epitope identification. However, when low-levels HLA/peptide tetramer-positive T cell populations are encountered, it is difficult to ascertain whether this represents a true T cell receptor (TCR)-mediated interaction or background signal. To address this issue, we have developed a method for both HLA class I and class II tetramer assays to confirm tetramer-binding to the TCR/CD3 complex. Preincubation of T cells with anti-CD3 mAb SPV-T3b and subsequent crosslinking interferes with the binding of HLA/peptide tetramers to the TCR/CD3 complex and thereby indicates to what extent HLA/peptide tetramer binds through interaction with TCR/CD3 complex. SPV-T3b pretreatment results in a 2- to 10-fold decrease in tetramer-binding intensity to antigen-specific CD8+ or CD4+ T cells, whereas background reactivity of HLA/peptide tetramers containing HIV-derived peptide in HIV-negative donors remained unchanged. SPV-T3b pretreatment forms a valuable tool to verify tetramer-based detection of antigen-specific T cells during the monitoring of immune responses in clinical studies.
Collapse
Affiliation(s)
- Pauline Weder
- Division of Immunology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Ton N M Schumacher
- Division of Immunology, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Hergen Spits
- Tytgat Institute for Liver and Instestinal Research, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Rosalie M Luiten
- Dept. of Dermatology and The Netherlands Institute for Pigment Disorders, Academic Medical Center, University of Amsterdam, The Netherlands
| |
Collapse
|
16
|
Laing KJ, Dong L, Sidney J, Sette A, Koelle DM. Immunology in the Clinic Review Series; focus on host responses: T cell responses to herpes simplex viruses. Clin Exp Immunol 2012; 167:47-58. [PMID: 22132884 PMCID: PMC3248086 DOI: 10.1111/j.1365-2249.2011.04502.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2011] [Indexed: 01/04/2023] Open
Abstract
Herpes virus infections are chronic and co-exist with acquired immune responses that generally prevent severe damage to the host, while allowing periodic shedding of virus and maintenance of its transmission in the community. Herpes simplex viruses type 1 and 2 (HSV-1, HSV-2) are typical in this regard and are representative of the viral subfamily Alphaherpesvirinae, which has a tropism for neuronal and epithelial cells. This review will emphasize recent progress in decoding the physiologically important CD8(+) and CD4(+) T cell responses to HSV in humans. The expanding data set is discussed in the context of the search for an effective HSV vaccine as therapy for existing infections and to prevent new infections.
Collapse
Affiliation(s)
- K J Laing
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
17
|
Wald A, Koelle DM, Fife K, Warren T, Leclair K, Chicz RM, Monks S, Levey DL, Musselli C, Srivastava PK. Safety and immunogenicity of long HSV-2 peptides complexed with rhHsc70 in HSV-2 seropositive persons. Vaccine 2011; 29:8520-9. [PMID: 21945262 DOI: 10.1016/j.vaccine.2011.09.046] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 09/07/2011] [Accepted: 09/12/2011] [Indexed: 12/27/2022]
Abstract
HSV-2, the primary causative agent of genital herpes, establishes latency in sensory ganglia and reactivates causing recurrent lesions and viral shedding. Induction or expansion of CD4(+) and CD8(+) T cell responses are expected to be important for a successful therapeutic vaccine against HSV-2. A candidate vaccine consisting of 32 synthetic 35mer HSV-2 peptides non-covalently complexed with recombinant human Hsc70 protein (named HerpV, formerly AG-707) was tested for safety and immunogenicity in a Phase I study. These peptides are derived from 22 HSV-2 proteins representative of all phases of viral replication. Thirty-five HSV-2 infected participants were randomized and treated in one of four groups: HerpV+QS-21 (saponin adjuvant), HerpV, QS-21, or vehicle. The vaccine was well tolerated and safe. All seven participants with evaluable samples who were administered HerpV with QS-21 demonstrated a statistically significant CD4(+) T cell response to HSV-2 antigens, and the majority of such participants demonstrated a statistically significant CD8(+) T cell response as well. To our knowledge, this is the first candidate vaccine against HSV-2 to demonstrate a broad CD4(+) and CD8(+) T cell response in HSV-2(+) participants, and the first HSP-based vaccine to show immune responses against viral antigens in humans.
Collapse
Affiliation(s)
- Anna Wald
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Use of HLA-DR*08032/E7 and HLA-DR*0818/E7 tetramers in tracking of epitope-specific CD4+ T cells in active and convalescent tuberculosis patients compared with control donors. Immunobiology 2011; 216:947-60. [PMID: 21281984 DOI: 10.1016/j.imbio.2011.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Revised: 01/02/2011] [Accepted: 01/04/2011] [Indexed: 11/22/2022]
Abstract
Comparative tracking of tetramer-positive and epitope-specific CD4(+) T cells in blood and other tissues from tuberculosis (TB) patients during TB development and treatment using control donor samples is not well characterized. In this study, a novel HLA-DR-restricted peptide E7 from the ESAT-6 protein of Mycobacterium tuberculosis (MTB) was used to prepare modified HLA-DR*08032/E7 tetramer (tetramer 1) and HLA-DR*0818/E7 tetramer (tetramer 2) to monitor a series of samples from TB patients and control donors. Tetramer staining showed that (1) by direct staining of single sample and flow cytometric analyses, detection of tetramer-positive CD4(+) T cells ranged from 0.1% to 8.8% (median 0.67% in tetramer 1 and 0.5% in tetramer 2), 0.1 to 10.7% (0.74% and 0.71%), 0.02 to 2.2% (0.25% and 0.25%), 0.02 to 0.48% (0.2% and 0.2%) and most at under 0-0.2% (0.2% and 0.16%) in the initial pulmonary TB (PTB) patients' blood, pleural fluid (PLF) of initial tuberculous pleuritis patients, non-TB patients' blood, healthy donors' blood and umbilical cord blood, respectively; significantly higher levels of CD4(+) T cells were detected in samples of TB patients than in three control donor groups; (2) by direct staining of time point TB samples and flow cytometric analyses, along with TB symptom amendment at day 60, tetramer-positive CD4(+) T cells began to decrease, until after 90-120 days, reached and kept at a relatively low even normal level about at 0.03-0.3%; (3) by enrichment approach, at least 10-fold increased memory tetramer-positive CD4(+) T cells were seen; (4) by in situ staining, tetramer-positive, IFN-γ-producing and/or TNF-α-producing CD4(+) T cells in the lymph node and lung granuloma and cavernous tissues of TB patients could be determined. Therefore, by further increasing the sample size tested to confirm the specificity and sensitivity of tetrameric molecules, it should be possible to develop them for use as research and diagnostic reagents.
Collapse
|
19
|
Ayyoub M, Dojcinovic D, Pignon P, Raimbaud I, Schmidt J, Luescher I, Valmori D. Monitoring of NY-ESO-1 specific CD4+ T cells using molecularly defined MHC class II/His-tag-peptide tetramers. Proc Natl Acad Sci U S A 2010; 107:7437-42. [PMID: 20368442 PMCID: PMC2867704 DOI: 10.1073/pnas.1001322107] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
MHC-peptide tetramers have become essential tools for T-cell analysis, but few MHC class II tetramers incorporating peptides from human tumor and self-antigens have been developed. Among limiting factors are the high polymorphism of class II molecules and the low binding capacity of the peptides. Here, we report the generation of molecularly defined tetramers using His-tagged peptides and isolation of folded MHC/peptide monomers by affinity purification. Using this strategy we generated tetramers of DR52b (DRB3*0202), an allele expressed by approximately half of Caucasians, incorporating an epitope from the tumor antigen NY-ESO-1. Molecularly defined tetramers avidly and stably bound to specific CD4(+) T cells with negligible background on nonspecific cells. Using molecularly defined DR52b/NY-ESO-1 tetramers, we could demonstrate that in DR52b(+) cancer patients immunized with a recombinant NY-ESO-1 vaccine, vaccine-induced tetramer-positive cells represent ex vivo in average 1:5,000 circulating CD4(+) T cells, include central and transitional memory polyfunctional populations, and do not include CD4(+)CD25(+)CD127(-) regulatory T cells. This approach may significantly accelerate the development of reliable MHC class II tetramers to monitor immune responses to tumor and self-antigens.
Collapse
Affiliation(s)
- Maha Ayyoub
- Institut National de la Santé et de la Recherche Médicale, Unité 892, Centre de Lutte Contre le Cancer René Gauducheau, 44800 Saint Herblain, France
| | - Danijel Dojcinovic
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066 Epalinges, Switzerland; and
| | - Pascale Pignon
- Institut National de la Santé et de la Recherche Médicale, Unité 892, Centre de Lutte Contre le Cancer René Gauducheau, 44800 Saint Herblain, France
| | - Isabelle Raimbaud
- Institut National de la Santé et de la Recherche Médicale, Unité 892, Centre de Lutte Contre le Cancer René Gauducheau, 44800 Saint Herblain, France
| | - Julien Schmidt
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066 Epalinges, Switzerland; and
| | - Immanuel Luescher
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, CH-1066 Epalinges, Switzerland; and
| | - Danila Valmori
- Institut National de la Santé et de la Recherche Médicale, Unité 892, Centre de Lutte Contre le Cancer René Gauducheau, 44800 Saint Herblain, France
- Faculty of Medicine, University of Nantes, 44093 Nantes, France
| |
Collapse
|
20
|
Wei H, Wang R, Yuan Z, Chen CY, Huang D, Halliday L, Zhong W, Zeng G, Shen Y, Shen L, Wang Y, Chen ZW. DR*W201/P65 tetramer visualization of epitope-specific CD4 T-cell during M. tuberculosis infection and its resting memory pool after BCG vaccination. PLoS One 2009; 4:e6905. [PMID: 19730727 PMCID: PMC2731856 DOI: 10.1371/journal.pone.0006905] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 08/07/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In vivo kinetics and frequencies of epitope-specific CD4 T cells in lymphoid compartments during M. tuberculosis infection and their resting memory pool after BCG vaccination remain unknown. METHODOLOGY/FINDINGS Macaque DR*W201 tetramer loaded with Ag85B peptide 65 was developed to directly measure epitope-specific CD4 T cells in blood and tissues form macaques after M. tuberculosis infection or BCG vaccination via direct staining and tetramer-enriched approach. The tetramer-based enrichment approach showed that P65 epitope-specific CD4 T cells emerged at mean frequencies of approximately 500 and approximately 4500 per 10(7) PBL at days 28 and 42, respectively, and at day 63 increased further to approximately 22,000/10(7) PBL after M. tuberculosis infection. Direct tetramer staining showed that the tetramer-bound P65-specific T cells constituted about 0.2-0.3% of CD4 T cells in PBL, lymph nodes, spleens, and lungs at day 63 post-infection. 10-fold expansion of these tetramer-bound epitope-specific CD4 T cells was seen after the P65 peptide stimulation of PBL and tissue lymphocytes. The tetramer-based enrichment approach detected BCG-elicited resting memory P65-specific CD4 T cells at a mean frequency of 2,700 per 10(7) PBL. SIGNIFICANCE Our work represents the first elucidation of in vivo kinetics and frequencies for tetramer-bound epitope-specific CD4 T cells in the blood, lymphoid tissues and lungs over times after M. tuberculosis infection, and BCG immunization.
Collapse
Affiliation(s)
- Huiyong Wei
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Richard Wang
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Zhuqing Yuan
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Crystal Y. Chen
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Dan Huang
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Lisa Halliday
- Biological Resource Laboratory, University of Illinois at Chicago (UIC), Chicago, Illinois, United States of America
| | - Weihua Zhong
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Gucheng Zeng
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Yun Shen
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Ling Shen
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Yunqi Wang
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
| | - Zheng W. Chen
- Department of Immunology & Microbiology, Center for Primate Biomedical Research, University of Illinois College of Medicine at Chicago (UIC), Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
21
|
Justesen S, Harndahl M, Lamberth K, Nielsen LLB, Buus S. Functional recombinant MHC class II molecules and high-throughput peptide-binding assays. Immunome Res 2009; 5:2. [PMID: 19416502 PMCID: PMC2690590 DOI: 10.1186/1745-7580-5-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 05/05/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Molecules of the class II major histocompability complex (MHC-II) specifically bind and present exogenously derived peptide epitopes to CD4+ T helper cells. The extreme polymorphism of the MHC-II hampers the complete analysis of peptide binding. It is also a significant hurdle in the generation of MHC-II molecules as reagents to study and manipulate specific T helper cell responses. Methods to generate functional MHC-II molecules recombinantly, and measure their interaction with peptides, would be highly desirable; however, no consensus methodology has yet emerged. RESULTS We generated alpha and beta MHC-II chain constructs, where the membrane-spanning regions were replaced by dimerization motifs, and the C-terminal of the beta chains was fused to a biotinylation signal peptide (BSP) allowing for in vivo biotinylation. These chains were produced separately as inclusion bodies in E. coli , extracted into urea, and purified under denaturing and non-reducing conditions using conventional column chromatography. Subsequently, diluting the two chains into a folding reaction with appropriate peptide resulted in efficient peptide-MHC-II complex formation. Several different formats of peptide-binding assay were developed including a homogeneous, non-radioactive, high-throughput (HTS) binding assay. Binding isotherms were generated allowing the affinities of interaction to be determined. The affinities of the best binders were found to be in the low nanomolar range. Recombinant MHC-II molecules and accompanying HTS peptide-binding assay were successfully developed for nine different MHC-II molecules including the DPA1*0103/DPB1*0401 (DP401) and DQA1*0501/DQB1*0201, where both alpha and beta chains are polymorphic, illustrating the advantages of producing the two chains separately. CONCLUSION We have successfully developed versatile MHC-II resources, which may assist in the generation of MHC class II -wide reagents, data, and tools.
Collapse
Affiliation(s)
- Sune Justesen
- Laboratory of Experimental Immunology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | |
Collapse
|
22
|
Cecconi V, Moro M, Del Mare S, Dellabona P, Casorati G. Use of MHC class II tetramers to investigate CD4+T cell responses: Problems and solutions. Cytometry A 2008; 73:1010-8. [DOI: 10.1002/cyto.a.20603] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
23
|
Vollers SS, Stern LJ. Class II major histocompatibility complex tetramer staining: progress, problems, and prospects. Immunology 2008; 123:305-13. [PMID: 18251991 DOI: 10.1111/j.1365-2567.2007.02801.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The use of major histocompatibility complex (MHC) tetramers in the detection and analysis of antigen-specific T cells has become more widespread since its introduction 11 years ago. Early challenges in the application of tetramer staining to CD4+ T cells centred around difficulties in the expression of various class II MHC allelic variants and the detection of low-frequency T cells in mixed populations. As many of the technical obstacles to class II MHC tetramer staining have been overcome, the focus has returned to uncertainties concerning how oligomer valency and T-cell receptor/MHC affinity affect tetramer binding. Such issues have become more important with an increase in the number of studies relying on direct ex vivo analysis of antigen-specific CD4+ T cells. In this review we discuss which problems in class II MHC tetramer staining have been solved to date, and which matters remain to be considered.
Collapse
Affiliation(s)
- Sabrina S Vollers
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | | |
Collapse
|
24
|
Wambre E, Van Overtvelt L, Maillère B, Humphreys R, von Hofe E, Ferhat L, Ebo D, Moingeon P. Single Cell Assessment of Allergen-Specific T Cell Responses with MHC Class II Peptide Tetramers: Methodological Aspects. Int Arch Allergy Immunol 2008; 146:99-112. [DOI: 10.1159/000113513] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Accepted: 10/16/2007] [Indexed: 12/14/2022] Open
|
25
|
Höhn H, Kortsik C, Zehbe I, Hitzler WE, Kayser K, Freitag K, Neukirch C, Andersen P, Doherty TM, Maeurer M. MHC class II tetramer guided detection of Mycobacterium tuberculosis-specific CD4+ T cells in peripheral blood from patients with pulmonary tuberculosis. Scand J Immunol 2007; 65:467-78. [PMID: 17444958 DOI: 10.1111/j.1365-3083.2007.01924.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Novel diagnostic tools are needed to diagnose latent infection and to provide biologically meaningful surrogate markers to define cellular immune responses against Mycobacterium tuberculosis (MTB). Interferon gamma-based assays have recently been developed in addition to the more than 100-year-old tuberculin skin test (TST) for the immune diagnosis of MTB in blood. The advent of soluble MHC/peptide tetramer molecules allows to objectively enumerate antigen-specific T cells. We identified novel MHC class II-restricted MTB epitopes and used HLA-DR4 tetrameric complexes to visualize ex vivo CD4(+) T cells directed against the antigens Ag85B and the 19-kDa lipoprotein, shared between MTB and other Mycobacterium species, and CD4(+) T cells which recognize the MTB-associated ESAT-6 antigen. MTB-reactive CD4(+) T cells reside predominantly in the CD45RA(+) CD28(+) and CD45(-) CD28(+) T-cell subset and recognize naturally processed and presented MTB epitopes. HLA-DR4-restricted, Ag85B or ESAT-6-specific CD4(+) T cells show similar dynamics over time in peripheral blood mononuclear cells (PBMC) when compared with CD8(+) T cells directed against the corresponding HLA-A2-presented MTB epitopes in patients with pulmonary MTB infection and subsequent successful therapy. This was not found to be true for T-cell responses directed against the 19-kDa lipoprotein. The dissection of the cellular immune response in M. tuberculosis infection will enable novel strategies for monitoring MTB vaccine candidates and to gauge CD4(+) T cells directed against MTB.
Collapse
Affiliation(s)
- H Höhn
- Department of Medical Microbiology, University of Mainz, Hochhaus Augustusplatz, Mainz, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Veldman C, Eming R, Wolff-Franke S, Sonderstrup G, Kwok WW, Hertl M. Detection of low avidity desmoglein 3-reactive T cells in pemphigus vulgaris using HLA-DR beta 1*0402 tetramers. Clin Immunol 2006; 122:330-7. [PMID: 17113829 DOI: 10.1016/j.clim.2006.09.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 09/14/2006] [Accepted: 09/23/2006] [Indexed: 11/21/2022]
Abstract
In the present study, we developed a HLA class II tetramer-based detection system utilizing DRB1*0402 tetramers loaded with recently identified immunodominant peptides of desmoglein 3 (Dsg3), the major autoantigen of pemphigus vulgaris (PV). Initial experiments demonstrated staining of a Dsg3-reactive T cell hybridoma which was derived from HLA-DR0402-transgenic mice with loaded PE-labeled DRbeta1*0402 tetramers. However, staining of autoreactive T cell clones (TCC) derived from PV patients resulted only in positive staining by addition of exogenous peptides to the staining reactions. There was a dose-dependent specific binding of TCC to the tetramers with the agonistic Dsg3 peptide which was not altered by exogenous unrelated Dsg3 peptide. Noteworthy, the TCC did not stain with HLA-DR4 tetramers complexed with unrelated Dsg3 peptides. The findings of this study suggest that HLA class II tetramers may provide a highly specific approach to monitor ex vivo the T cellular autoimmune response against Dsg3 in patients with PV.
Collapse
Affiliation(s)
- Christian Veldman
- Department of Dermatology and Allergology, University of Marburg, Deutschhausstrasse 9, D-35033 Marburg, Germany.
| | | | | | | | | | | |
Collapse
|
27
|
Bioley G, Jandus C, Tuyaerts S, Rimoldi D, Kwok WW, Speiser DE, Tiercy JM, Thielemans K, Cerottini JC, Romero P. Melan-A/MART-1-Specific CD4 T Cells in Melanoma Patients: Identification of New Epitopes and Ex Vivo Visualization of Specific T Cells by MHC Class II Tetramers. THE JOURNAL OF IMMUNOLOGY 2006; 177:6769-79. [PMID: 17082590 DOI: 10.4049/jimmunol.177.10.6769] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the past decade, many efforts have been made to identify MHC class II-restricted epitopes from different tumor-associated Ags. Melan-A/MART-1(26-35) parental or Melan-A/MART-1(26-35(A27L)) analog epitopes have been widely used in melanoma immunotherapy to induce and boost CTL responses, but only one Th epitope is currently known (Melan-A51-73, DRB1*0401 restricted). In this study, we describe two novel Melan-A/MART-1-derived sequences recognized by CD4 T cells from melanoma patients. These epitopes can be mimicked by peptides Melan-A27-40 presented by HLA-DRB1*0101 and HLA-DRB1*0102 and Melan-A25-36 presented by HLA-DQB1*0602 and HLA-DRB1*0301. CD4 T cell clones specific for these epitopes recognize Melan-A/MART-1+ tumor cells and Melan-A/MART-1-transduced EBV-B cells and recognition is reduced by inhibitors of the MHC class II presentation pathway. This suggests that the epitopes are naturally processed and presented by EBV-B cells and melanoma cells. Moreover, Melan-A-specific Abs could be detected in the serum of patients with measurable CD4 T cell responses specific for Melan-A/MART-1. Interestingly, even the short Melan-A/MART-1(26-35(A27L)) peptide was recognized by CD4 T cells from HLA-DQ6+ and HLA-DR3+ melanoma patients. Using Melan-A/MART-1(25-36)/DQ6 tetramers, we could detect Ag-specific CD4 T cells directly ex vivo in circulating lymphocytes of a melanoma patient. Together, these results provide the basis for monitoring of naturally occurring and vaccine-induced Melan-A/MART-1-specific CD4 T cell responses, allowing precise and ex vivo characterization of responding T cells.
Collapse
Affiliation(s)
- Gilles Bioley
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, University Hospital, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Koelle DM, Huang J, Hensel MT, McClurkan CL. Innate immune responses to herpes simplex virus type 2 influence skin homing molecule expression by memory CD4+ lymphocytes. J Virol 2006; 80:2863-72. [PMID: 16501095 PMCID: PMC1395438 DOI: 10.1128/jvi.80.6.2863-2872.2006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Herpes simplex virus (HSV) infections of humans are characterized by intermittent, lytic replication in epithelia. Circulating HSV-specific CD4 T cells express lower levels of preformed cutaneous lymphocyte-associated antigen (CLA), a skin-homing receptor, than do circulating HSV-specific CD8 T cells but, paradoxically, move into infected skin earlier than CD8 cells. Memory CD4 T cells develop strong and selective expression of CLA and E-selectin ligand while responding to HSV antigen in vitro. We now show that interleukin-12, type I interferon, and transforming growth factor beta are each involved in CLA expression by memory HSV type 2 (HSV-2)-specific CD4 T cells in peripheral blood mononuclear cells (PBMC). A reduction of the number of monocytes and dendritic cells from PBMC reduces CLA expression by HSV-2-responsive CD4 lymphoblasts, while their reintroduction restores this phenotype, identifying these cells as possible sources of CLA-promoting cytokines. Plasmacytoid dendritic cells are particularly potent inducers of CLA on HSV-reactive CD4 T cells. These observations are consistent with cooperation between innate and acquired immunity to promote a pattern of homing receptor expression that is physiologically appropriate for trafficking to infected tissues.
Collapse
Affiliation(s)
- David M Koelle
- Department of Medicine, University of Washington, Seattle, USA.
| | | | | | | |
Collapse
|
29
|
Reichstetter S, Standifer NE, Geubtner KA, Liu AW, Agar SL, Kwok WW. Cytotoxic herpes simplex type 2-specific, DQ0602-restricted CD4 T+-cell clones show alloreactivity to DQ0601. Immunology 2006; 117:350-7. [PMID: 16476054 PMCID: PMC1782233 DOI: 10.1111/j.1365-2567.2005.02308.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Alloreactivity is one of the most serious problems in organ transplantation. It has been hypothesized that pre-existing alloreactive T cells are actually cross-reacting cells that have been primed by the autologous major histocompatibility complex (MHC) and a specific peptide. CD8+ cytotoxic T lymphocytes that are alloreactive and recognize a virus-peptide that is presented by the autologous MHC have been reported. Here we demonstrate a cross-reactivity that exists between DQ0602 restricted, herpes simplex type 2 VP16 40-50 specific CD4+ T-cell clones, which can be alloreactive to DQ0601. Though most of the DQ0602 restricted T-cell clones we isolated from two different donors were not alloreactive, weakly cross-reacting T-cell clones could be isolated from both donors. Two strongly cross-reacting T-cell clones with high affinity interaction of their T-cell receptor (TCR) with both DQ0602/VP16 40-50 and DQ0601 could be isolated from one donor. DNA sequencing of the a fragment of the Vbeta gene used in their TCR confirmed that these two T cells indeed are two independent clones. These clones are cytotoxic and produce cytokines of a T helper 2-like pattern. Possible implications in a DR-matched transplantation setting are discussed.
Collapse
|
30
|
Abstract
Recognition of a peptide-MHC complex by the T cell receptor (TCR) is a key interaction that initiates T lymphocyte activation or silencing during an immune response. Fluorochrome-labeled recombinant MHC class II-peptide reagents function as soluble mimetics of this interaction, bind to their specific TCR, and allow for detection of antigen-specific CD4+ T cells. These reagents are now under scrutiny for "immune staging" of patients at risk of type 1 diabetes, in an effort to diagnose islet autoimmunity early enough to block immune-mediated beta cell destruction. Several issues are currently being addressed to improve the performance of these T cell assays: enrichment steps for better sensitivity, multiplexing of several islet epitopes, simultaneous monitoring of CD4+ and CD8+ responses, detection of low avidity T cells, combination of quantitative (number of positive cells) and qualitative (cytokine secretion, naive/memory phenotype) readouts. CD4+ T cells are key effectors of autoimmunity, and these MHC class II peptide reagents, through their signaling properties, might also provide therapeutics to block the autoimmune process at its onset, analogous to the use of OKT3gammao1(AlaAla) anti-CD3 antibody but in an antigen-specific fashion. The aim of such therapeutics is to potentiate different physiological control mechanisms to restore immune tolerance. Mechanisms initiated by this pathway may be capable of triggering elimination of pathogenic T cells through antigen-specific apoptosis and anergy, combined with the induction of regulatory T cells with broad suppressive function.
Collapse
Affiliation(s)
- Roberto Mallone
- Benaroya Research Institute at Virginia Mason and Department of Immunology
University of Washington School of MedicineUS
| | - Gerald T. Nepom
- Benaroya Research Institute at Virginia Mason and Department of Immunology
University of Washington School of MedicineUS
| |
Collapse
|
31
|
Norimine J, Brown WC. Intrahaplotype and interhaplotype pairing of bovine leukocyte antigen DQA and DQB molecules generate functional DQ molecules important for priming CD4+ T-lymphocyte responses. Immunogenetics 2005; 57:750-62. [PMID: 16220347 DOI: 10.1007/s00251-005-0045-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Accepted: 08/10/2005] [Indexed: 10/25/2022]
Abstract
Antigen-specific CD4(+) T-lymphocyte responses are restricted by major histocompatibility complex class II molecules, which influence T-cell priming during infection. Human leukocyte antigen (HLA) and bovine leukocyte antigen (BoLA) DRB3 and DQ genes are polymorphic, but unlike HLA, many BoLA haplotypes have duplicated DQ genes, and antibody-blocking studies indicated that BoLA-DQ molecules present various pathogen epitopes. Limited experimentation also suggested that BoLA-DQ molecules formed by interhaplotype pairing of A and B chains are functional. To compare antigen presentation by DR and DQ molecules and to definitively demonstrate functional BoLA-DQ molecules derived from interhaplotype pairing, different combinations of DR or DQ A and B proteins were expressed with CD80 in 293-F cells for use as antigen-presenting cells (APCs). This approach identified 11 unique restriction elements including five DR and six DQ pairs for antigen-specific CD4(+) T-cell responses against tick-transmitted bovine hemoparasites Anaplasma marginale or Babesia bovis. Interhaplotype pairing of DQ A and B molecules was demonstrated. Testing of six expressed DQA/B pairs from an animal with duplicated DQ haplotypes (DH16A/DH22H) demonstrated that an interhaplotype pair, DQA*2206/DQB*1301, presented A. marginale peptide B. In DH22H and DH16A homozygous animals, DQA*2206 was tightly linked with DQB*1402, and DQA*22021 was linked with DQB*1301. APCs from these donors could not present peptide B, confirming that DQA*2206/DQB*1301 encoded a functional interhaplotype pair. Functional BoLA-DQ molecules are generated by both intrahaplotype and interhaplotype pairing of A and B chains and play a similar role to BoLA-DR in priming helper T-cell responses to important pathogens.
Collapse
Affiliation(s)
- Junzo Norimine
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164-7040, USA.
| | | |
Collapse
|
32
|
Kinnunen T, Kwok WW, Närvänen A, Rytkönen-Nissinen M, Immonen A, Saarelainen S, Taivainen A, Virtanen T. Immunomodulatory potential of heteroclitic analogs of the dominant T-cell epitope of lipocalin allergen Bos d 2 on specific T cells. Int Immunol 2005; 17:1573-81. [PMID: 16221722 DOI: 10.1093/intimm/dxh332] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Peptide-based allergen immunotherapy is a novel alternative for conventional allergen immunotherapy. Here, we have characterized the immunomodulatory potential of heteroclitic peptide analogs of the immunodominant epitope of lipocalin allergen Bos d 2 on specific human T-cell clones. The TCR affinity of Bos d 2-specific T-cell clones for the natural peptide ligand and its heteroclitic analogs was assessed with fluorescent-labeled MHC class II tetramers. The activation and cytokine production of the clones were analyzed upon stimulation with the different ligands. Moreover, the capacity of the heteroclitic analogs to induce hyporesponsiveness and cell death was examined. The T-cell clones F1-9 and K3-2 bound MHC class II tetramers loaded with the heteroclitic peptide analogs of the immunodominant epitope of Bos d 2 with increased affinity. At similar peptide concentrations, stimulation of the clones with the heteroclitic analogs favored increased IFN-gamma/IL-4 and IFN-gamma/IL-5 ratios in comparison with stimulation with the natural peptide ligand. Moreover, the T-cell clones stimulated with the heteroclitic analogs exhibited an increased susceptibility to cell death or hyporesponsiveness upon re-stimulation. Our results suggest that heteroclitic analogs of a T-cell epitope of an allergen may enhance the efficacy of peptide-based allergen immunotherapy.
Collapse
Affiliation(s)
- Tuure Kinnunen
- Department of Clinical Microbiology, University of Kuopio, PO Box 1627, FIN-70211 Kuopio, Finland.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Niemiec PK, Read LR, Sharif S. Synthesis of chicken major histocompatibility complex class II oligomers using a baculovirus expression system. Protein Expr Purif 2005; 46:390-400. [PMID: 16236525 DOI: 10.1016/j.pep.2005.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 08/29/2005] [Accepted: 09/01/2005] [Indexed: 11/15/2022]
Abstract
Chicken major histocompatibility complex (MHC) B21 and B19 haplotypes are associated with resistance and susceptibility to Marek's disease (MD), respectively. T-cell-mediated immune response is crucial in coordinating protection against Marek's disease virus (MDV) infection, but it has been difficult to identify and characterize antigen-specific T-cells. MHC class II tetramers and oligomers have been widely used for characterization of antigen-specific T-cells in the context of infectious and autoimmune diseases. Thus, the objective of this study was to synthesize chicken MHC class II oligomers of B21 and B19 haplotypes for the future identification of antigen-specific T-cells. To achieve this objective, full-length coding sequences of chicken MHC class II B21 and B19 molecules were amplified and the molecules were expressed as fusion proteins, carrying Fos and Jun leucine zipper (LZ), histidine-tag and biotin ligase recognition site sequences, using a baculovirus expression system. Recombinant MHC-II were loaded with self-peptides, which stabilized the heterodimer in SDS-PAGE and allowed the detection of these molecules in Western blots with a conformation-specific anti-chicken MHC class II antibody. Biotinylated MHC molecules were conjugated to streptavidin to form oligomers, which were resolved under the transmission electron microscope through immuno-gold labelling, thus confirming success of oligomerization. In conclusion, chicken MHC class II oligomers may be used in the future to study the antigen-specific CD4+ T-cell compartment.
Collapse
Affiliation(s)
- Patrycja K Niemiec
- Department of Pathobiology, University of Guelph, Guelph, Ont., Canada N1G 2W1
| | | | | |
Collapse
|
34
|
Abstract
PROBLEM Genital herpes simplex infections are generally limited to epithelia and neurons. Vaccines have had activity in herpes simplex virus (HSV)-seronegative women only. Understanding how HSV-specific T cells traffic to infected sites may assist in vaccine design. METHOD OF STUDY Herpes simplex virus epitopes recognized by HSV-specific CD8 T cells were identified and used to make fluorescent human leukocyte antigen (HLA)-peptide tetramers. Molecules related to lymphocyte rolling adhesion were studied by flow cytometry and cell binding. HSV-specific CD4 T cells identified ex vivo by cytokine accumulation or activation marker expression, or detected in vitro by 5-(and-6)-carboxyfluorescein diacetate, succinimidyl ester (CFSE) dilution, were similarly investigated. RESULTS Herpes simplex virus-specific T cells are 10- to 100-fold more prevalent in lesional skin compared with blood and greatly enriched in lesions compared with normal skin. Diverse viral antigens are recognized by HSV-specific T cells. Functionally active E-selectin ligand, and cutaneous lymphocyte antigen (CLA), are expressed by circulating HSV-2-specific CD8 cells. CD4 cells display lower levels of CLA that are dramatically up-regulated upon re-stimulation with antigen. CONCLUSIONS Herpes simplex virus-2-specific CD8 and CD4 T cells differ in constitutive expression of skin homing molecules. Vaccines designed to induce proper homing are postulated to have increased efficacy.
Collapse
Affiliation(s)
- David M Koelle
- Department of Medicine, University of Washington, Seattle, WA 98104, USA.
| | | | | |
Collapse
|
35
|
Danke NA, Koelle DM, Kwok WW. Persistence of Herpes Simplex Virus Type 2 VP16-Specific CD4+ T Cells. Hum Immunol 2005; 66:777-87. [PMID: 16112025 DOI: 10.1016/j.humimm.2005.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Revised: 03/11/2005] [Accepted: 03/14/2005] [Indexed: 10/25/2022]
Abstract
Patients with genital herpes have frequent viral reactivations. The repeated antigenic rechallenges can modulate the CD4+ memory T-cell repertoires during the course of infection. In this study, the CD4+ T-cell responses against the herpes simplex virus type 2 (HSV-2) tegument protein VP16 were studied in two HSV-2-infected subjects at two different time points that spanned a 5-year period. Although the VP16-specific T cells did exhibit variation of T-cell receptor Vbeta usages at the two time points, T cells that used identical Vbeta and CDR3 junction sequences were also observed at the two time points. These experiments demonstrate that the CD4+ T cells that are directed against HSV-2 VP16 protein in chronically infected individuals are oligoclonal and that T cells of specific clonotypes can be maintained throughout the course of the disease.
Collapse
Affiliation(s)
- Nancy A Danke
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | | | |
Collapse
|
36
|
Zhang Y, Renkvist N, Sun Z, Schuler-Thurner B, Glaichenhaus N, Schuler G, Boon T, van der Bruggen P, Colau D. A polyclonal anti-vaccine CD4 T cell response detected with HLA-DP4 multimers in a melanoma patient vaccinated with MAGE-3.DP4-peptide-pulsed dendritic cells. Eur J Immunol 2005; 35:1066-75. [PMID: 15756643 DOI: 10.1002/eji.200425847] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During the last few years, HLA class I tetramers have been successfully used to demonstrate anti-vaccine CD8 CTL proliferation in cancer patients vaccinated with tumor antigens. Frequencies of CTL as low as 10(-6) among CD8 cells were observed even in patients showing tumor regression. Little is known about the role of tumor-antigen-specific CD4 T cells in the context of these anti-vaccine responses. Therefore, we developed a very sensitive approach using fluorescent class-II-peptide multimers to detect antigen-specific CD4 T cells in vaccinated cancer patients. We produced HLA-DP4 multimers loaded with the MAGE-3(243-258) peptide and used them to stain ex vivo PBL from melanoma patients injected with dendritic cells pulsed with several class I and class II tumor antigenic peptides, including the MAGE-3(243-258) peptide. The multimer(+) CD4 T cells were sorted and amplified in clonal conditions; specificity was assessed by their ability to secrete IFN-gamma upon contact with the MAGE-3 antigen. We detected frequencies of about 1x10(-6) anti-MAGE-3.DP4 cells among CD4 cells. A detailed analysis of one patient showed an anti-MAGE-3.DP4 CD4 T cell amplification of at least 3000-fold upon immunization. TCR analysis of the clones from this patient demonstrated a polyclonal response against the MAGE-3 peptide.
Collapse
Affiliation(s)
- Yi Zhang
- Ludwig Institute for Cancer Research and Cellular Genetics Unit, Université de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chaves FA, Hou P, Wu S, Sant AJ. Replacement of the membrane proximal region of I-Ad MHC class II molecule with I-E-derived sequences promotes production of an active and stable soluble heterodimer without altering peptide-binding specificity. J Immunol Methods 2005; 300:74-92. [PMID: 15896797 DOI: 10.1016/j.jim.2005.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Revised: 02/28/2005] [Accepted: 02/28/2005] [Indexed: 11/24/2022]
Abstract
The MHC class II molecule I-A is the murine homologue of HLA-DQ in humans. The I-A and DQ heterodimers display considerable heterodimer instability compared with their I-E and HLA-DR counterparts. This isotype-specific behavior makes the production of soluble I-A and DQ molecules very difficult. We have developed a strategy for production of soluble I-A(d) molecules involving expression of I-A(d) as a glycosil phosphatidyl inositol (PI) anchored chimera in Chinese Hamster Ovary (CHO) cells. The regions comprising the membrane proximal segments of I-A(d) alpha and beta chains were substituted for the corresponding regions of I-E, and the derived constructs were expressed in CHO cells. Procedures for purification of the soluble class II molecules were optimized and the WT and chimeric molecule were compared for structure, biochemical stability and functionality. Our analysis revealed that the substitutions in the membrane proximal domains improved cell surface expression and thermal stability of I-A(d) without altering the peptide binding specificity of the class II molecule. The results suggest that similar strategies could be used to increase the stability of other unstable class II molecules for in vitro studies.
Collapse
Affiliation(s)
- Francisco A Chaves
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
38
|
Lacey SF, Diamond DJ, Zaia JA. Assessment of cellular immunity to human cytomegalovirus in recipients of allogeneic stem cell transplants. Biol Blood Marrow Transplant 2005; 10:433-47. [PMID: 15205665 DOI: 10.1016/j.bbmt.2003.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Effective reconstitution of cellular immunity following hematopoietic stem cell transplantation (HCT) is thought to be important for protection from the morbidity caused by cytomegalovirus (CMV) reactivation and disease. This review critically discusses current methods for assessment of CMV-specific cellular immune responses, with emphasis on flow cytometry-based methodologies such as MHC-I and MHC-II tetramer staining and intracellular cytokine assays. The advantages and weaknesses of these assays are considered in comparison to traditional immunologic techniques. Application of these newer methodologies has provided insight into the dynamics of the levels of CMV-specific CD4(+) and CD8(+) T-lymphocytes following HCT, and into the sources and diversity of these cells. Data from preliminary clinical studies suggest that CMV-specific CD8(+) T-lymphocyte levels greater than 1 x 10(7)/L of peripheral blood may correlate with protection from CMV disease. Studies on the functional phenotypes of CMV-specific CD8(+) T-lymphocytes such as cytokine production, degranulation, and cytotoxicity have indicated that these cells are heterogeneous with regard to these properties. Future research will focus on establishing whether any of these immunologic assays will serve as a correlate of protection and inform as to which patients are at high risk for CMV reactivation and disease. Identification of an informative assay may allow its incorporation into standard clinical practice for monitoring HCT patients.
Collapse
Affiliation(s)
- Simon F Lacey
- Laboratory of Vaccine Research, Beckman Research Institute of the City of Hope, Duarte, California 91010-3000, USA.
| | | | | |
Collapse
|
39
|
Giannoni F, Barnett J, Bi K, Samodal R, Lanza P, Marchese P, Billetta R, Vita R, Klein MR, Prakken B, Kwok WW, Sercarz E, Altman A, Albani S. Clustering of T cell ligands on artificial APC membranes influences T cell activation and protein kinase C theta translocation to the T cell plasma membrane. THE JOURNAL OF IMMUNOLOGY 2005; 174:3204-11. [PMID: 15749850 DOI: 10.4049/jimmunol.174.6.3204] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
T cell activation is associated with active clustering of relevant molecules in membrane microdomains defined as the supramolecular activation cluster. The contact area between these regions on the surface of T cells and APC is defined as the immunological synapse. It has been recently shown that preclustering of MHC-peptide complexes in membrane microdomains on the APC surface affects the efficiency of immune synapse formation and the related T cell activation. Disruption of such clusters may reduce the efficiency of stimulation. We describe here an entirely artificial system for Ag-specific, ex vivo stimulation of human polyclonal T cells (artificial APC (aAPC)). aAPC are based on artificial membrane bilayers containing discrete membrane microdomains encompassing T cell ligands (i.e., appropriate MHC-peptide complexes in association with costimulatory molecules). We show here that preclustering of T cell ligands triggered a degree of T cell activation significantly higher than the one achieved when we used either soluble tetramers or aAPC in which MHC-peptide complexes were uniformly distributed within artificial bilayer membranes. This increased efficiency in stimulation was mirrored by increased translocation from the cytoplasm to the membrane of protein kinase theta, a T cell signaling molecule that colocalizes with the TCR within the supramolecular activation cluster, thus indicating efficient engagement of T cell activation pathways. Engineered aAPC may have immediate application for basic and clinical immunology studies pertaining to modulation of T cells ex vivo.
Collapse
Affiliation(s)
- Francesca Giannoni
- Department of Medicine, University of California, San Diego, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zimmermann VS, Benigni F, Mondino A. Immune surveillance and anti-tumor immune responses: an anatomical perspective. Immunol Lett 2005; 98:1-8. [PMID: 15790503 DOI: 10.1016/j.imlet.2004.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2004] [Revised: 09/06/2004] [Accepted: 09/10/2004] [Indexed: 11/15/2022]
Abstract
The development of adaptive immune responses against infectious agents relies on the initiation of antigen specific immune responses in secondary lymphoid organs and on the migration of effector cells at the site of infection. Similarly, the development of anti-tumor immunity depends on the recognition of tumor-derived antigens by specific lymphocytes in the context of the lymphoid tissues and on the re-localisation of the cells to the site of cell transformation. Here, we will review the preclinical studies, which have defined the spatial and temporal organisation of anti-tumor immunity, and discuss the implications of these findings in active immunotherapy.
Collapse
Affiliation(s)
- Valerie S Zimmermann
- Cancer Immunotherapy and Gene Therapy Program, San Raffaele Scientific Institute, DIBIT, Via Olgettina 58, 20132 Milan, Italy
| | | | | |
Collapse
|
41
|
Nesburn AB, Ramos TV, Zhu X, Asgarzadeh H, Nguyen V, BenMohamed L. Local and systemic B cell and Th1 responses induced following ocular mucosal delivery of multiple epitopes of herpes simplex virus type 1 glycoprotein D together with cytosine-phosphate-guanine adjuvant. Vaccine 2005; 23:873-83. [PMID: 15603887 DOI: 10.1016/j.vaccine.2004.08.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Revised: 06/21/2004] [Accepted: 08/05/2004] [Indexed: 11/17/2022]
Abstract
Vaccine strategies that stimulate the ocular mucosal immune system (OMIS), the immune barrier that protects the surface of the eye are needed. However, most vaccines fail to induce local ocular immune responses and, in the absence of adjuvant, may induce a state of immunological tolerance. In this study, we present a new vaccine strategy that consists of ocular mucosal (OM) delivery of peptide epitopes, selected from the herpes simplex virus (HSV-1) glycoprotein D (gD) mixed with synthetic immunostimulatory oligodeoxynucleotides (ODNs) containing unmethylated CpG motifs (CpG2007). Repeated topical ocular application of gD peptide epitopes and CpG2007 induced peptide-specific and virus-neutralizing IgA/IgG in tears as well as in serum. As a second marker, generation of local and systemic peptide- and virus-specific T cells confirmed the potent immunogenicity of peptides-CpG2007 formulation when applied through the OM route. Moreover, OM delivery of peptides-CpG2007 induced local IFN-gamma and IL-2 responses and low IL-4 production, demonstrating the polarization towards a Th1 response. Immunization, using free CpG2007 ODNs or peptides alone did not produce OMIS stimulation. This novel vaccine strategy may be key for ocular infectious pathogens, such as HSV-1, that require both secretory antibody and the Th1 responses. The results suggest the clinical feasibility of developing an OM delivery system using epitope-based vaccines.
Collapse
Affiliation(s)
- Anthony B Nesburn
- Laboratory of Cellular and Molecular Immunology, Department of Ophthalmology, College of Medicine, University of California at Irvine, Irvine, CA 92697-4375, USA
| | | | | | | | | | | |
Collapse
|
42
|
González JC, Kwok WW, Wald A, McClurkan CL, Huang J, Koelle DM. Expression of cutaneous lymphocyte-associated antigen and E-selectin ligand by circulating human memory CD4+ T lymphocytes specific for herpes simplex virus type 2. J Infect Dis 2004; 191:243-54. [PMID: 15609235 PMCID: PMC1255909 DOI: 10.1086/426944] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Accepted: 08/09/2004] [Indexed: 01/26/2023] Open
Abstract
Virus-specific memory T lymphocytes traffic to sites of viral infection. Herpes simplex virus (HSV) type 2-specific CD4(+) and CD8(+) T lymphocytes differ with regard to their homing kinetics to infected tissues. We studied the expression of cutaneous lymphocyte-associated antigen (CLA) and E-selectin ligand (ESL) by HSV-2-specific CD4(+) T lymphocytes. Virus-reactive T lymphocytes were identified ex vivo by CD154 or interferon-gamma up-regulation. We detected selective expression of CLA by HSV-2-reactive CD4(+) T lymphocytes, but at levels lower than those we previously observed for CD8(+) T lymphocytes. Short-term HSV-2-reactive CD4(+) lines generated from peripheral-blood mononuclear cells preferentially express CLA, compared with cytomegalovirus- or influenza-specific cells. CLA is expressed by HSV-2-reactive cells that are initially CLA negative before restimulation. Short-term culture-expanded HSV-2-specific CD4(+) T lymphocytes also selectively express ESL. These findings have implications for the optimization of vaccines for HSV and other cutaneous pathogens.
Collapse
Affiliation(s)
| | | | - Anna Wald
- Departments of Medicine
- Epidemiology, and
| | | | - Jay Huang
- Laboratory Medicine, University of Washington
| | - David M. Koelle
- Departments of Medicine
- Pathobiology
- Laboratory Medicine, University of Washington
- Fred Hutchinson Cancer Research Center, and
- Benaroya Research Institute, Seattle, Washington
- Reprints or correspondence: Dr. David M. Koelle, Harborview Medical Center, Box 359690, 325 Ninth Ave., Seattle, WA 98104 (
)
| |
Collapse
|
43
|
Mohindru M, Kang B, Kim BS. Functional maturation of proteolipid protein(139-151)-specific Th1 cells in the central nervous system in experimental autoimmune encephalomyelitis. J Neuroimmunol 2004; 155:127-35. [PMID: 15342203 DOI: 10.1016/j.jneuroim.2004.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2004] [Revised: 06/28/2004] [Accepted: 06/28/2004] [Indexed: 10/26/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a widely adopted animal model system for studying human multiple sclerosis that affects the central nervous system (CNS). To understand the underlying pathogenic mechanisms of the autoimmune T cell response, localization, enumeration and characterization of autoreactive T cells are essential. We assessed encephalitogenic proteolipid protein epitope (PLP(139-151))-specific T cells in the periphery and CNS of SJL/J mice using MHC class II I-As multimers during both pre-clinical and clinical phases of PLP-induced EAE in conjunction with T cell function. Our results strongly suggest that PLP(139-151)-specific CD4+ T cells first expand primarily in the CNS-draining cervical lymph nodes and then migrate to the CNS. In the CNS, these PLP-specific CD4+ T cells accumulate, become activated and differentiate into effector cells that produce IFN-gamma in response to the self-peptide.
Collapse
Affiliation(s)
- Mani Mohindru
- Department of Microbiology-Immunology and Institute for Neuroscience, Northwestern University Feinberg Medical School, 303 East Chicago Ave, Chicago, IL 60611, USA
| | | | | |
Collapse
|
44
|
Affiliation(s)
- J W Gratama
- Laboratory for Clinical and Tumor Immunology, Department of Medical Oncology, Erasmus MC-Daniel den Hoed, Rotterdam, the Netherlands.
| | | |
Collapse
|
45
|
Yang J, Jaramillo A, Shi R, Kwok WW, Mohanakumar T. In vivo biotinylation of the major histocompatibility complex (MHC) class II/peptide complex by coexpression of BirA enzyme for the generation of MHC class II/tetramers. Hum Immunol 2004; 65:692-9. [PMID: 15301857 DOI: 10.1016/j.humimm.2004.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Success in generation of major histocompatibility complex (MHC) tetramer relies on application of a key technique, biotinylation of MHC molecule specifically on a single lysine residue using the BirA enzyme. However, in vitro biotinylation of MHC-BSP (BirA enzyme substrate peptide) fusion protein using BirA enzyme is laborious and is prone to losses of target proteins to unacceptable levels. To circumvent this problem, an in vivo biotinylation strategy was developed where the BirA enzyme was coexpressed with target protein, HLA-DR2BSP/MBP, in an insect cell expression system. Bacterial BirA enzyme expressed in Drosophila melanogaster 2 (D. Mel-2) cell lines was biologically functional and was able to biotinylate secretary target protein (on specific lysine residue present on the BSP tag). Biotinylation efficiency was maximized by providing exogenous d-biotin in the culture medium and optimization of the expression vector ratios for cotransfection. By limiting dilution cloning, a clone was identified where the expressed DR2BSP/MBP protein was completely biotinylated. DR2BSP/MBP protein expressed and purified from such a clone was ready to be tetramerized with streptavidin to be used for staining antigen-specific T cells.
Collapse
Affiliation(s)
- Junbao Yang
- Department of Surgery, Washington University School of Medicine, MO 63110-1093, USA
| | | | | | | | | |
Collapse
|
46
|
Huang MMS, Borszcz P, Sidobre S, Kronenberg M, Kane KP. CD1d1 Displayed on Cell Size Beads Identifies and Enriches an NK Cell Population Negatively Regulated by CD1d1. THE JOURNAL OF IMMUNOLOGY 2004; 172:5304-12. [PMID: 15100269 DOI: 10.4049/jimmunol.172.9.5304] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NK cells destroy microbe-infected cells while sparing healthy cells, and are controlled, in part, by inhibitory receptors specific for class I Ag-presenting molecules. CD1d1, a beta(2)-microglobulin-associated class I-like molecule, binds glycolipids and stimulates NKT cells. We previously demonstrated that target cell lysis by IL-2-activated mouse NK cells is inhibited by target cell expression of CD1d1, suggesting that IL-2-activated NK cells may express a CD1d1-specific inhibitory receptor. We now report that a significant subset of mouse IL-2-activated NK cells specifically binds cell size beads displaying either naturally expressed or recombinant CD1d1. In contrast, although tetramers of soluble recombinant CD1d1 loaded with alpha-galactosylceramide identify NKT cells, binding of this reagent to resting or IL-2-activated NK cells was undetectable, even with activated NK cells sorted with CD1d1 beads. Cytotoxicity by the CD1d1 bead-separated NK subset was strongly inhibited by CD1d1, compared with the NK cell subset not bound to CD1d1 beads. An Ab that blocks NKT cell recognition of CD1d1 also reverses CD1d1 inhibition of NK lysis, suggesting that TCRs of NKT cells and NK inhibitory receptor(s) may interact with a similar site on CD1d1. These results provide direct evidence for a physical interaction of NK cells with CD1d1, mediated by a functional, CD1d1-specific low-affinity inhibitory NK receptor. Display of ligands on cell size beads to maximize multivalent interaction may offer an alternative approach to examine NK cell receptor-ligand interactions, particularly those of lower expression and/or lower affinity/avidity that may go undetected using tetrameric reagents.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Antigens, CD1/biosynthesis
- Antigens, CD1/immunology
- Antigens, CD1/metabolism
- Antigens, CD1/physiology
- Antigens, CD1d
- Cell Communication/immunology
- Cell Line, Tumor
- Cell Size
- Cells, Cultured
- Cytotoxicity, Immunologic/immunology
- Down-Regulation/immunology
- Female
- Galactosylceramides/metabolism
- Immunosuppressive Agents/immunology
- Immunosuppressive Agents/metabolism
- Immunosuppressive Agents/pharmacology
- Interleukin-2/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/immunology
- Lymphocyte Subsets/cytology
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microspheres
- Protein Binding/immunology
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/physiology
- Receptors, KIR
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Michael M S Huang
- Department of Medical Microbiology and Immunology, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
47
|
Mallone R, Nepom GT. MHC Class II tetramers and the pursuit of antigen-specific T cells: define, deviate, delete. Clin Immunol 2004; 110:232-42. [PMID: 15047201 DOI: 10.1016/j.clim.2003.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Accepted: 11/10/2003] [Indexed: 10/26/2022]
Abstract
Selective expansion and activation of a very small number of antigen-specific CD4(+) T cells is a remarkable and essential property of the adaptive immune response. Antigen-specific T cells were until recently identified only indirectly by functional assays, such as antigen-induced cytokine secretion and proliferation. The advent of MHC Class II tetramers has added a pivotal tool to our research armamentarium, allowing the definition of allo- and autoimmune responses in deeper detail. Rare antigen-specific CD4(+) cells can now be selectively identified, isolated and characterized. The same tetramer reagents also provide a new mean of stimulating T cells, more closely reproducing the MHC-peptide/TCR interaction. This property allows the use of tetramers to direct T cells toward the more desirable outcome, that is, activation (in malignancies and infectious diseases) or Th2/T regulatory cell deviation, anergy and deletion (in autoimmune diseases). These experimental approaches hold promise for diagnostic, prognostic and therapeutic applications.
Collapse
Affiliation(s)
- Roberto Mallone
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA.
| | | |
Collapse
|
48
|
Landais E, Saulquin X, Scotet E, Trautmann L, Peyrat MA, Yates JL, Kwok WW, Bonneville M, Houssaint E. Direct killing of Epstein-Barr virus (EBV)–infected B cells by CD4 T cells directed against the EBV lytic protein BHRF1. Blood 2004; 103:1408-16. [PMID: 14563644 DOI: 10.1182/blood-2003-03-0930] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Due to their low frequency, CD4 T-cell responses to Epstein-Barr virus (EBV) lytic antigens are, so far, poorly characterized. Human peptide major histocompatibility complex (MHC) class II multimers provide a means to detect and characterize such rare T cells. Along a screening of T-cell responses to lytic or latent EBV antigens within peripheral blood leukocyte (PBL)– or synovial-derived CD4 T-cell lines, we identified an human leukocyte antigen–DR*0401 (HLA-DR*0401)–restricted epitope derived from BHRF1 (BamHI fragment H rightward open reading frame 1), a viral protein produced during the early stages of the lytic cycle. We show here that T-cell responses to this particular BHRF1 epitope are shared by most EBV-infected DR*0401+ individuals, as BHRF1-specific CD4 T cells could be sorted out from all the DRB*0401 T-cell lines analyzed, using magnetic beads coated with recombinant BHRF1/DR*0401 complexes. Sorting with these peptide MHC class II multimers was very efficient, as the yield of recovery of BHRF1-specific T cells was nearly 100%. Functional analysis of a large number of clones responding to BHRF1/DR*0401 demonstrated their cytolytic action against autologous and allogeneic DR*0401+ EBV-transformed B-lymphoblastoid cell lines (B-LCLs), with 40% to 80% killing efficiency and potent interferon γ production, thus suggesting that this CD4 T-cell population contributes to the control of EBV replication. B-LCL lysis by these T-cell clones was DR*0401 dependent, EBV dependent, and was not merely due to bystander killing. Taken together, these data provide the first demonstration that a lytic antigen can induce a direct cytolytic response against EBV-infected cells.
Collapse
Affiliation(s)
- Elise Landais
- INSERM U463, Institut de Biologie, 9 quai Moncousu, 44035 Nantes Cedex 01, France
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The development of reliable in vitro assays that could allow the quantitation and characterization of anti-donor alloimmune responses has always been a goal in clinical transplantation, both to predict presensitization to the transplanted tissue and to be able to identify rejection without resorting to more invasive tests. With recent development in our understanding of transplantation biology and therapeutics, there is a real expectation that these tests may be used to identify tolerance as much as to predict rejection. The traditional limiting dilution assays still have a contribution to make and are being complemented by an array of tools, such as ELISpot, flow cytometry-based techniques, and microarray analysis. The assays that have been informative, to date, are discussed in this review. This information will lead, at least, to a better understanding of how and when the rejection process occurs. More interestingly, the objective is to apply this information to evaluate tolerance-inducing strategies or to identify patients that have become tolerant to their graft and can be weaned of immunosuppression. Of course sensitive, accurate and specific immunologic monitoring has applications well beyond the field of transplantation.
Collapse
Affiliation(s)
- Maria P Hernandez-Fuentes
- Department of Immunology, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | | | | |
Collapse
|
50
|
Mannering SI, Morris JS, Jensen KP, Purcell AW, Honeyman MC, van Endert PM, Harrison LC. A sensitive method for detecting proliferation of rare autoantigen-specific human T cells. J Immunol Methods 2003; 283:173-83. [PMID: 14659909 DOI: 10.1016/j.jim.2003.09.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The ability to measure proliferation of rare antigen-specific T cells among many bystanders is critical for the evaluation of cellular immune function in health and disease. T-cell proliferation in response to antigen has been measured almost exclusively by 3H-thymidine incorporation. This method does not directly identify the phenotype of the proliferating cells and is frequently not sufficiently sensitive to detect rare autoantigen-specific T cells. To overcome these problems, we developed a novel assay for antigen-specific human T-cell proliferation. Peripheral blood mononuclear cells (PBMC) were labelled with the fluorescent dye 5,6-carboxylfluorescein diacetate succinimidyl ester (CFSE) and cells that proliferated in response to antigen, with resultant reduction in CFSE intensity, were measured directly by flow cytometry. This assay was more sensitive than 3H-thymidine incorporation and detected the proliferation of rare antigen-specific CD4(+) T cells at 10-fold lower antigen concentrations. It also allowed the phenotype of the proliferating cells to be directly determined. Using the CFSE assay we were able to measure directly the proliferation of human CD4(+) T cells from healthy donors in response to the type 1 diabetes autoantigens glutamic acid decarboxylase (GAD) and proinsulin (PI).
Collapse
Affiliation(s)
- Stuart I Mannering
- Autoimmunity and Transplantation Division, The Walter and Eliza Hall Institute, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| | | | | | | | | | | | | |
Collapse
|