1
|
Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology 2021; 1:1557-1576. [PMID: 23264902 PMCID: PMC3525611 DOI: 10.4161/onci.22428] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prophylactic vaccination constitutes one of the most prominent medical achievements of history. This concept was first demonstrated by the pioneer work of Edward Jenner, dating back to the late 1790s, after which an array of preparations that confer life-long protective immunity against several infectious agents has been developed. The ensuing implementation of nation-wide vaccination programs has de facto abated the incidence of dreadful diseases including rabies, typhoid, cholera and many others. Among all, the most impressive result of vaccination campaigns is surely represented by the eradication of natural smallpox infection, which was definitively certified by the WHO in 1980. The idea of employing vaccines as anticancer interventions was first theorized in the 1890s by Paul Ehrlich and William Coley. However, it soon became clear that while vaccination could be efficiently employed as a preventive measure against infectious agents, anticancer vaccines would have to (1) operate as therapeutic, rather than preventive, interventions (at least in the vast majority of settings), and (2) circumvent the fact that tumor cells often fail to elicit immune responses. During the past 30 y, along with the recognition that the immune system is not irresponsive to tumors (as it was initially thought) and that malignant cells express tumor-associated antigens whereby they can be discriminated from normal cells, considerable efforts have been dedicated to the development of anticancer vaccines. Some of these approaches, encompassing cell-based, DNA-based and purified component-based preparations, have already been shown to exert conspicuous anticancer effects in cohorts of patients affected by both hematological and solid malignancies. In this Trial Watch, we will summarize the results of recent clinical trials that have evaluated/are evaluating purified peptides or full-length proteins as therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Teixeira AG, Kleinman A, Agarwal R, Tam NW, Wang J, Frampton JP. Confinement of Suspension-Cultured Cells in Polyethylene Glycol/Polyethylene Oxide-Albumin Aqueous Two-Phase Systems. Front Chem 2019; 7:441. [PMID: 31275925 PMCID: PMC6591268 DOI: 10.3389/fchem.2019.00441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 05/28/2019] [Indexed: 11/13/2022] Open
Abstract
Aqueous two-phase systems (ATPSs) have numerous applications in separation science, and more recently, in bioassays enabled by the solution micropatterning of cells. The most frequently used ATPS in these applications is the polyethylene glycol (PEG)-dextran (Dex) system, as the polymers that form this ATPS have been extensively characterized in terms of their physicochemical properties. However, in addition to this well-known system, there exist many other ATPSs with properties that may be exploited to improve upon the PEG-dextran system for specific applications. One of these underexplored systems is the ATPS formed from PEG/polyethylene oxide (PEO) and albumin. In this article, we characterize the phase separation of PEG (35 kDa) and polyethylene oxide (PEO) (200, 900, and 4,000 kDa) with bovine serum albumin (BSA). We describe the microscopic emulsion behavior of these systems in the presence of NaCl and compounds (NaHCO3, NaH2PO4, and HEPES) commonly used in buffer solutions and cell culture media. We further demonstrate that PEG- and PEO-albumin systems can be used in place of the PEG-dextran system for confinement of suspension-cultured cells (Jurkat T cells and RPMI-8226 B cells). Cell viability and morphology are examined for various polymer formulations relative to the commonly used PEG 35 kDa-Dex 500 kDa system and polymer-free cell culture medium. In addition, we examine cell activation for various phase-separating medium components by measuring IL-2 and IL-6 secretion. We demonstrate that we can confine immune cells and cytokines in the PEG-BSA system, and that this system can be employed to screen immune responses by enzyme-linked immunospot (ELISpot) assay. This new system represents a promising ATPS formulation for applications where low levels of baseline cell activation are required, for instance, when culturing immune cells.
Collapse
Affiliation(s)
- Alyne G. Teixeira
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | | | - Rishima Agarwal
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | - Nicky W. Tam
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | - Jun Wang
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Canadian Center for Vaccinology, IWK Health Centre, Halifax, NS, Canada
| | - John P. Frampton
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
3
|
Dey S, Kamil Reza K, Wuethrich A, Korbie D, Ibn Sina AA, Trau M. Tracking antigen specific T-cells: Technological advancement and limitations. Biotechnol Adv 2018; 37:145-153. [PMID: 30508573 DOI: 10.1016/j.biotechadv.2018.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/30/2018] [Accepted: 11/20/2018] [Indexed: 11/18/2022]
Abstract
Assessing T-cell mediated immune status can help to understand the body's response to disease and also provide essential diagnostic information. However, detection and characterization of immune response are challenging due to the rarity of signature biomolecules in biological fluid and require highly sensitive and specific assay technique for the analysis. Until now, several techniques spanning from flow cytometry to microsensors have been developed or under investigation for T-cell mediated immune response monitoring. Most of the current assays are designed to estimate average immune responses, i.e., total functional protein analysis and detection of total T-cells irrespective of their antigen specificity. Although potential, immune response analysis without detecting and characterizing the rare subset of T-cell population could lead to over or underestimation of patient's immune status. Addressing this limitation, recently a number of technological advancements in biosensing have been developed for this. The potential of simple and precise micro-technologies including microarray and microfluidic platforms for assessing antigen-specific T-cells will be highlighted in this review, together with a discussion on existing challenges and future aspects of immune-sensor development.
Collapse
Affiliation(s)
- Shuvashis Dey
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - K Kamil Reza
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Alain Wuethrich
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Darren Korbie
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Abu Ali Ibn Sina
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia.
| | - Matt Trau
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia; School of Chemistry and Molecular Biosciences, The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
4
|
Shen C, Xu T, Wu Y, Li X, Xia L, Wang W, Shahzad KA, Zhang L, Wan X, Qiu J. Frequency and reactivity of antigen-specific T cells were concurrently measured through the combination of artificial antigen-presenting cell, MACS and ELISPOT. Sci Rep 2017; 7:16400. [PMID: 29180767 PMCID: PMC5703716 DOI: 10.1038/s41598-017-16549-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 11/14/2017] [Indexed: 01/24/2023] Open
Abstract
Conventional peptide-major histocompatibility complex (pMHC) multimer staining, intracellular cytokine staining, and enzyme-linked immunospot (ELISPOT) assay cannot concurrently determine the frequency and reactivity of antigen-specific T cells (AST) in a single assay. In this report, pMHC multimer, magnetic-activated cell sorting (MACS), and ELISPOT techniques have been integrated into a micro well by coupling pMHC multimers onto cell-sized magnetic beads to characterize AST cell populations in a 96-well microplate which pre-coated with cytokine-capture antibodies. This method, termed AAPC-microplate, allows the enumeration and local cytokine production of AST cells in a single assay without using flow cytometry or fluorescence intensity scanning, thus will be widely applicable. Here, ovalbumin257-264-specific CD8+ T cells from OT-1 T cell receptor (TCR) transgenic mice were measured. The methodological accuracy, specificity, reproducibility, and sensitivity in enumerating AST cells compared well with conventional pMHC multimer staining. Furthermore, the AAPC-microplate was applied to detect the frequency and reactivity of Hepatitis B virus (HBV) core antigen18-27- and surface antigen183-191-specific CD8+ T cells for the patients, and was compared with conventional method. This method without the need of high-end instruments may facilitate the routine analysis of patient-specific cellular immune response pattern to a given antigen in translational studies.
Collapse
Affiliation(s)
- Chuanlai Shen
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China.
| | - Tao Xu
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - You Wu
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Xiaoe Li
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Lingzhi Xia
- Department of Laboratory Medicine, Nanjing KingMed Diagnostics Company Limited, Nanjing, Jiangsu, China
| | - Wei Wang
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Khawar Ali Shahzad
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Lei Zhang
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Xin Wan
- Department of Microbiology and Immunology, Southeast University Medical School, Nanjing, Jiangsu, China
| | - Jie Qiu
- Division of Infectious Diseases, Second Hospital of Nanjing, Affiliated Second Hospital of Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Bentzen AK, Hadrup SR. Evolution of MHC-based technologies used for detection of antigen-responsive T cells. Cancer Immunol Immunother 2017; 66:657-666. [PMID: 28314956 PMCID: PMC5406421 DOI: 10.1007/s00262-017-1971-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 02/03/2017] [Indexed: 01/07/2023]
Abstract
T cell-mediated recognition of peptide-major histocompatibility complex (pMHC) class I and II molecules is crucial for the control of intracellular pathogens and cancer, as well as for stimulation and maintenance of efficient cytotoxic responses. Such interactions may also play a role in the development of autoimmune diseases. Novel insights into this mechanism are crucial to understanding disease development and establishing new treatment strategies. MHC multimers have been used for detection of antigen-responsive T cells since the first report by Altman et al. showed that tetramerization of pMHC class I molecules provided sufficient stability to T cell receptor (TCR)-pMHC interactions, allowing detection of MHC multimer-binding T cells using flow cytometry. Since this breakthrough the scientific community has aimed for expanding the capacity of MHC multimer-based detection technologies to facilitate large-scale epitope discovery and immune monitoring in limited biological material. Screening of T cell specificity using large libraries of pMHC molecules is suitable for analyses of T cell recognition potentially at genome-wide levels rather than analyses restricted to a selection of model antigens. Such strategies provide novel insights into the immune specificities involved in disease development and response to immunotherapy, and extend fundamental knowledge related to T cell recognition patterns and cross-recognition by TCRs. MHC multimer-based technologies have now evolved from detection of 1-2 different T cell specificities per cell sample, to include more than 1000 evaluable pMHC molecules using novel technologies. Here, we provide an overview of MHC multimer-based detection technologies developed over two decades, focusing primarily on MHC class I interactions.
Collapse
Affiliation(s)
- Amalie Kai Bentzen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark.
| |
Collapse
|
6
|
A Simple Microfluidic Platform for Long-Term Analysis and Continuous Dual-Imaging Detection of T-Cell Secreted IFN-γ and IL-2 on Antibody-Based Biochip. BIOSENSORS-BASEL 2015; 5:750-67. [PMID: 26690235 PMCID: PMC4697143 DOI: 10.3390/bios5040750] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/12/2015] [Accepted: 11/25/2015] [Indexed: 01/11/2023]
Abstract
The identification and characterization, at the cellular level, of cytokine productions present a high interest for both fundamental research and clinical studies. However, the majority of techniques currently available (ELISA, ELISpot, flow cytometry, etc.) have several shortcomings including, notably, the assessment of several cytokines in relation to individual secreting cells and the monitoring of living cell responses for a long incubation time. In the present work, we describe a system composed of a microfluidic platform coupled with an antibody microarray chip for continuous SPR imaging and immunofluorescence analysis of cytokines (IL-2 and IFN-γ) secreted by T-Lymphocytes, specifically, and stably captured on the biochip under flow upon continued long-term on-chip culture (more than 24 h).
Collapse
|
7
|
Brooks SE, Bonney SA, Lee C, Publicover A, Khan G, Smits EL, Sigurdardottir D, Arno M, Li D, Mills KI, Pulford K, Banham AH, van Tendeloo V, Mufti GJ, Rammensee HG, Elliott TJ, Orchard KH, Guinn BA. Application of the pMHC Array to Characterise Tumour Antigen Specific T Cell Populations in Leukaemia Patients at Disease Diagnosis. PLoS One 2015; 10:e0140483. [PMID: 26492414 PMCID: PMC4619595 DOI: 10.1371/journal.pone.0140483] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/25/2015] [Indexed: 01/03/2023] Open
Abstract
Immunotherapy treatments for cancer are becoming increasingly successful, however to further improve our understanding of the T-cell recognition involved in effective responses and to encourage moves towards the development of personalised treatments for leukaemia immunotherapy, precise antigenic targets in individual patients have been identified. Cellular arrays using peptide-MHC (pMHC) tetramers allow the simultaneous detection of different antigen specific T-cell populations naturally circulating in patients and normal donors. We have developed the pMHC array to detect CD8+ T-cell populations in leukaemia patients that recognise epitopes within viral antigens (cytomegalovirus (CMV) and influenza (Flu)) and leukaemia antigens (including Per Arnt Sim domain 1 (PASD1), MelanA, Wilms' Tumour (WT1) and tyrosinase). We show that the pMHC array is at least as sensitive as flow cytometry and has the potential to rapidly identify more than 40 specific T-cell populations in a small sample of T-cells (0.8-1.4 x 10(6)). Fourteen of the twenty-six acute myeloid leukaemia (AML) patients analysed had T cells that recognised tumour antigen epitopes, and eight of these recognised PASD1 epitopes. Other tumour epitopes recognised were MelanA (n = 3), tyrosinase (n = 3) and WT1(126-134) (n = 1). One of the seven acute lymphocytic leukaemia (ALL) patients analysed had T cells that recognised the MUC1(950-958) epitope. In the future the pMHC array may be used provide point of care T-cell analyses, predict patient response to conventional therapy and direct personalised immunotherapy for patients.
Collapse
MESH Headings
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antigens, Nuclear/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Cell Separation
- Epitopes/immunology
- Flow Cytometry
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/immunology
- Major Histocompatibility Complex/immunology
- Peptides/immunology
- Reproducibility of Results
Collapse
Affiliation(s)
- Suzanne E. Brooks
- Cancer Sciences Unit (MP824), Somers Cancer Sciences Building, University of Southampton, Southampton, United Kingdom
| | - Stephanie A. Bonney
- Cancer Sciences Unit (MP824), Somers Cancer Sciences Building, University of Southampton, Southampton, United Kingdom
| | - Cindy Lee
- Cancer Sciences Unit (MP824), Somers Cancer Sciences Building, University of Southampton, Southampton, United Kingdom
- Department of Haematology, Southampton University Hospitals Trust, University of Southampton, Southampton, United Kingdom
| | - Amy Publicover
- Department of Haematology, Southampton University Hospitals Trust, University of Southampton, Southampton, United Kingdom
| | - Ghazala Khan
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, United Kingdom
| | - Evelien L. Smits
- Laboratory of Experimental Haematology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijkstraat 10, B-2650 Antwerp, Belgium
| | - Dagmar Sigurdardottir
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Matthew Arno
- King’s Genomics Centre, School of Biomedical and Health Sciences, King's College London, London, United Kingdom
| | - Demin Li
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Ken I. Mills
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology (CCRCB), Queen’s University Belfast, Belfast, United Kingdom
| | - Karen Pulford
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Alison H. Banham
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Viggo van Tendeloo
- Laboratory of Experimental Haematology, Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijkstraat 10, B-2650 Antwerp, Belgium
| | - Ghulam J. Mufti
- Department of Haematological Medicine, King's College London School of Medicine, London, United Kingdom
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Tim J. Elliott
- Cancer Sciences Unit (MP824), Somers Cancer Sciences Building, University of Southampton, Southampton, United Kingdom
| | - Kim H. Orchard
- Department of Haematology, Southampton University Hospitals Trust, University of Southampton, Southampton, United Kingdom
| | - Barbara-ann Guinn
- Cancer Sciences Unit (MP824), Somers Cancer Sciences Building, University of Southampton, Southampton, United Kingdom
- Department of Life Sciences, University of Bedfordshire, Park Square, Luton, United Kingdom
- Department of Haematological Medicine, King's College London School of Medicine, London, United Kingdom
| |
Collapse
|
8
|
Wang X, Li S, Zhang P, Lv F, Liu L, Li L, Wang S. An optical nanoruler based on a conjugated polymer-silver nanoprism pair for label-free protein detection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:6040-6045. [PMID: 26314928 DOI: 10.1002/adma.201502880] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/29/2015] [Indexed: 06/04/2023]
Abstract
An optical nanoruler system based on a conjugated polyelectrolyte-silver nanoprism pair is developed for label-free protein detection by taking advantage of the metal-enhanced fluorescence effect of silver nanostructures. Antibody-antigen interactions induce a change in the metal-fluorophore distance, followed by the response of a fluorescent signal of the conjugated polyelectrolyte. The system is used to detect target antigens sensitively and selectively.
Collapse
Affiliation(s)
- Xiaoyu Wang
- State Key Laboratory for Advanced Metals and Materials, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
- Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Shengliang Li
- Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Pengbo Zhang
- Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Fengting Lv
- Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Libing Liu
- Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Lidong Li
- State Key Laboratory for Advanced Metals and Materials, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
| | - Shu Wang
- Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| |
Collapse
|
9
|
Christo SN, Diener KR, Hayball JD. The functional contribution of calcium ion flux heterogeneity in T cells. Immunol Cell Biol 2015; 93:694-704. [PMID: 25823995 DOI: 10.1038/icb.2015.34] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/15/2015] [Accepted: 02/16/2015] [Indexed: 12/30/2022]
Abstract
The role of intracellular calcium ion oscillations in T-cell physiology is being increasingly appreciated by studies that describe how unique temporal and spatial calcium ion signatures can control different signalling pathways. Within this review, we provide detailed mechanisms of calcium ion oscillations, and emphasise the pivotal role that calcium signalling plays in directing crucial events pertaining to T-cell functionality. We also describe methods of calcium ion quantification, and take the opportunity to discuss how a deeper understanding of calcium signalling combined with new detection and quantification methodologies can be used to better design immunotherapies targeting T-cell responses.
Collapse
Affiliation(s)
- Susan N Christo
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Kerrilyn R Diener
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia.,Robinson Research Institute, School of Paediatrics and Reproductive Health, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, Sansom Institute and Hanson Institute, School of Pharmacy and Medical Science, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
10
|
Dunston CR, Herbert R, Griffiths HR. Improving T cell-induced response to subunit vaccines: opportunities for a proteomic systems approach. ACTA ACUST UNITED AC 2015; 67:290-9. [PMID: 25708693 DOI: 10.1111/jphp.12383] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 11/23/2014] [Indexed: 11/30/2022]
Abstract
UNLABELLED Prophylactic vaccines are an effective strategy to prevent development of many infectious diseases. With new and re-emerging infections posing increasing risks to food stocks and the health of the population in general, there is a need to improve the rationale of vaccine development. One key challenge lies in development of an effective T cell-induced response to subunit vaccines at specific sites and in different populations. OBJECTIVES In this review, we consider how a proteomic systems-based approach can be used to identify putative novel vaccine targets, may be adopted to characterise subunit vaccines and adjuvants fully. KEY FINDINGS Despite the extensive potential for proteomics to aid our understanding of subunit vaccine nature, little work has been reported on identifying MHC 1-binding peptides for subunit vaccines generating T cell responses in the literature to date. SUMMARY In combination with predictive and structural biology approaches to mapping antigen presentation, proteomics offers a powerful and as yet un-tapped addition to the armoury of vaccine discovery to predict T-cell subset responses and improve vaccine design strategies.
Collapse
Affiliation(s)
- Christopher R Dunston
- Life & Health Sciences, Aston University, Birmingham, West Midlands, UK; Mologic, Bedford Technology Park, Thurleigh, Bedfordshire, MK44 2YP
| | | | | |
Collapse
|
11
|
Scrutinizing calcium flux oscillations in T lymphocytes to deduce the strength of stimulus. Sci Rep 2015; 5:7760. [PMID: 25585590 PMCID: PMC4293621 DOI: 10.1038/srep07760] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/11/2014] [Indexed: 01/13/2023] Open
Abstract
The capture and activation of individual T cells on functionalised surfaces enables real-time analyses of the magnitude and rhythm of intracellular calcium release. Application of Haarlet transformations generate a calcium flux ‘threshold’, with the frequency of the ‘threshold crossings’ correlating with the strength of the original T cell stimulus. These findings represent a new method to evaluate graduations in T cell activation in real time, and at a single-cell level.
Collapse
|
12
|
Bhavsar NA, Bream JH, Meeker AK, Drake CG, Peskoe SB, Dabitao D, De Marzo AM, Isaacs WB, Platz EA. A peripheral circulating TH1 cytokine profile is inversely associated with prostate cancer risk in CLUE II. Cancer Epidemiol Biomarkers Prev 2014; 23:2561-7. [PMID: 25150281 DOI: 10.1158/1055-9965.epi-14-0010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND TH1 cytokines, such as IFNγ and TNFα, and potentially innate cytokines, such as IL6, can potentiate the immune response to tumor. Cytokines, such as IL1β, IL8, and IL10, may suppress anticancer immunity. Thus, we prospectively evaluated the association between peripheral-cytokine concentrations and prostate cancer. METHODS We conducted an age-race matched case-control study (268 pairs) of incident prostate cancer in CLUE-II. We measured plasma IFNγ, IL10, IL12p70, IL1β, IL6, IL8, and TNFα concentrations using an ultrasensitive multiplex kit. ORs and 95% confidence intervals (CI) were calculated using conditional logistic regression. RESULTS The OR of prostate cancer decreased across quartiles of IFNγ (highest vs. lowest quartiles: OR, 0.49; 95% CI, 0.30-0.81; Ptrend = 0.006), TNFα (OR, 0.56; 95% CI, 0.33-0.96; Ptrend = 0.01), and IL6 (OR, 0.46; 95% CI, 0.26-0.79; Ptrend = 0.007). Higher TNFα (OR, 0.28; 95% CI, 0.09-0.85; Ptrend = 0.01) and IL6 (OR, 0.20; 95% CI, 0.06-0.67; Ptrend = 0.003) concentrations were associated with lower Gleason sum ≥7 disease risk. Other cytokines were not as clearly associated with risk. CONCLUSIONS Men with a prediagnostic circulating TH1 profile and higher IL6 may have a lower risk of prostate cancer, including aggressive disease. Whether this profile reflects (i) an intraprostatic immune environment in benign tissue that protects against prostate cancer, (ii) the immune milieu in response to a prostate adenocarcinoma that inhibits tumor growth and detectability, and/or (iii) a systemic immune profile that mediates the influence of modifiable factors on risk, warrants additional study. IMPACT Identifying specific inflammatory cytokines associated with prostate cancer may lead to improved prevention and treatment strategies.
Collapse
Affiliation(s)
- Nrupen A Bhavsar
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland. Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland. Center for Learning Health Care, Duke Clinical Research Institute, Durham, North Carolina
| | - Jay H Bream
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland. Department of Urology and James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Charles G Drake
- Department of Urology and James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland. Department of Immunology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sarah B Peskoe
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Djeneba Dabitao
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland. Department of Urology and James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - William B Isaacs
- Department of Urology and James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland. Department of Urology and James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland.
| |
Collapse
|
13
|
Kwak M, Kim DJ, Lee MR, Wu Y, Han L, Lee SK, Fan R. Nanowire array chips for molecular typing of rare trafficking leukocytes with application to neurodegenerative pathology. NANOSCALE 2014; 6:6537-50. [PMID: 24705924 PMCID: PMC4048658 DOI: 10.1039/c3nr06465d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Despite the presence of the blood-brain barrier (BBB) that restricts the entry of immune cells and mediators into the central nervous system (CNS), a small number of peripheral leukocytes can traverse the BBB and infiltrate into the CNS. The cerebrospinal fluid (CSF) is one of the major routes through which trafficking leukocytes migrate into the CNS. Therefore, the number of leukocytes and their phenotypic compositions in the CSF may represent important sources to investigate immune-to-brain interactions or diagnose and monitor neurodegenerative diseases. Due to the paucity of trafficking leucocytes in the CSF, a technology capable of efficient isolation, enumeration, and molecular typing of these cells in the clinical settings has not been achieved. In this study, we report on a biofunctionalized silicon nanowire array chip for highly efficient capture and multiplexed phenotyping of rare trafficking leukocytes in small quantities (50 microliters) of clinical CSF specimens collected from neurodegenerative disease patients. The antibody coated 3D nanostructured materials exhibited vastly improved rare cell capture efficiency due to high-affinity binding and enhanced cell-substrate interactions. Moreover, our platform creates multiple cell capture interfaces, each of which can selectively isolate specific leukocyte phenotypes. A comparison with the traditional immunophenotyping using flow cytometry demonstrated that our novel silicon nanowire-based rare cell analysis platform can perform rapid detection and simultaneous molecular characterization of heterogeneous immune cells. Multiplexed molecular typing of rare leukocytes in CSF samples collected from Alzheimer's disease patients revealed the elevation of white blood cell counts and significant alterations in the distribution of major leukocyte phenotypes. Our technology represents a practical tool for potentially diagnosing and monitoring the pathogenesis of neurodegenerative diseases by allowing an effective hematological analysis of the CSF from patients.
Collapse
Affiliation(s)
- Minsuk Kwak
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Zhao L, Zhang M, Cong H. Advances in the study of HLA-restricted epitope vaccines. Hum Vaccin Immunother 2013; 9:2566-77. [PMID: 23955319 DOI: 10.4161/hv.26088] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Vaccination is a proven strategy for protection from disease. An ideal vaccine would include antigens that elicit a safe and effective protective immune response. HLA-restricted epitope vaccines, which include T-lymphocyte epitopes restricted by HLA alleles, represent a new and promising immunization approach. In recent years, research in HLA-restricted epitope vaccines for the treatment of tumors and for the prevention of viral, bacterial, and parasite-induced infectious diseases have achieved substantial progress. Approaches for the improvement of the immunogenicity of epitope vaccines include (1) improving the accuracy of the methods used for the prediction of epitopes, (2) making use of additional HLA-restricted CD8(+) T-cell epitopes, (3) the inclusion of specific CD4(+) T-cell epitopes, (4) adding B-cell epitopes to the vaccine construction, (5) finding more effective adjuvants and delivery systems, (6) using immunogenic carrier proteins, and (7) using multiple proteins as epitopes sources. In this manuscript, we review recent research into HLA-restricted epitope vaccines.
Collapse
Affiliation(s)
- Lingxiao Zhao
- Department of Human Parasitology; Shandong University School of Medicine; Shandong, P.R. China
| | - Min Zhang
- Department of Human Parasitology; Shandong University School of Medicine; Shandong, P.R. China
| | - Hua Cong
- Department of Human Parasitology; Shandong University School of Medicine; Shandong, P.R. China
| |
Collapse
|
15
|
New targets for the immunotherapy of colon cancer-does reactive disease hold the answer? Cancer Gene Ther 2013; 20:157-68. [PMID: 23492821 DOI: 10.1038/cgt.2013.5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers in both men and women, posing a serious demographic and economic burden worldwide. In the United Kingdom, CRC affects 1 in every 20 people and it is often detected once well established and after it has spread beyond the bowel (Stage IIA-C and Stage IIIA-C). A diagnosis at such advanced stages is associated with poor treatment response and survival. However, studies have identified two sub-groups of post-treatment CRC patients--those with good outcome (reactive disease) and those with poor outcome (non-reactive disease). We aim to review the state-of-the-art for CRC with respect to the expression of cancer-testis antigens (CTAs) and their identification, evaluation and correlation with disease progression, treatment response and survival. We will also discuss the relationship between CTA expression and regulatory T-cell (Treg) activity to tumorigenesis and tumor immune evasion in CRC and how this could account for the clinical presentation of CRC. Understanding the molecular basis of reactive CRC may help us identify more potent novel immunotherapeutic targets to aid the effective treatment of this disease. In this review, based on our presentation at the 2012 International Society for the Cell and Gene Therapy of Cancer annual meeting, we will summarize some of the most current advances in CTA and CRC research and their influence on the development of novel immunotherapeutic approaches for this common and at times difficult to treat disease.
Collapse
|
16
|
Szmacinski H, Toshchakov V, Piao W, Lakowicz JR. Imaging of Protein Secretion from a Single Cell Using Plasmonic Substrates. BIONANOSCIENCE 2013; 3:30-36. [PMID: 23814699 PMCID: PMC3693482 DOI: 10.1007/s12668-013-0076-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Detecting, imaging, and monitoring cell function on a single cell basis is very important in the field of immunology research where many molecules are secreted from cells in response to external stimuli including immunization. Here we introduce substrates with plasmonic nanoparticles and fluorescence microscopy as promising imaging methods for studies on molecular processes controlling cell behavior, particularly secretion of cytokines. We developed unique composition of silver and silica layers of plasmonic nanostructures which resulted in fluorescence enhancement of more than 200-fold for ensemble of molecules in the immunoassay. For the proof of concept demonstration, we used primary mouse macrophages and imaged tumor necrosis alpha (TNF-α) secretion after stimulation of the cells with lipopolysaccharide (LPS). We demonstrate that metal-enhanced fluorescence assay provides imaging capability detection of cytokine secretion from a single cell without extensive biochemical procedures as required with standard methods. In addition it is demonstrated that cell viability can be controlled during secretion.
Collapse
Affiliation(s)
- Henryk Szmacinski
- Center for Fluorescence Spectroscopy, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 725 W. Lombard St., Baltimore, MD, 21201, USA
| | - Vladimir Toshchakov
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Redwood St., Baltimore, MD, 21201, USA
| | - Wenji Piao
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Redwood St., Baltimore, MD, 21201, USA
| | - Joseph R. Lakowicz
- Center for Fluorescence Spectroscopy, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 725 W. Lombard St., Baltimore, MD, 21201, USA
| |
Collapse
|
17
|
Birnbaum ME, Dong S, Garcia KC. Diversity-oriented approaches for interrogating T-cell receptor repertoire, ligand recognition, and function. Immunol Rev 2013; 250:82-101. [PMID: 23046124 DOI: 10.1111/imr.12006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Molecular diversity lies at the heart of adaptive immunity. T-cell receptors and peptide-major histocompatibility complex molecules utilize and rely upon an enormous degree of diversity at the levels of genetics, chemistry, and structure to engage one another and carry out their functions. This high level of diversity complicates the systematic study of important aspects of T-cell biology, but recent technical advances have allowed for the ability to study diversity in a comprehensive manner. In this review, we assess insights gained into T-cell receptor function and biology from our increasingly precise ability to assess the T-cell repertoire as a whole or to perturb individual receptors with engineered reagents. We conclude with a perspective on a new class of high-affinity, non-stimulatory peptide ligands we have recently discovered using diversity-oriented techniques that challenges notions for how we think about T-cell receptor signaling.
Collapse
Affiliation(s)
- Michael E Birnbaum
- Department of Molecular and Cellular Physiology, Program in Immunology, Stanford University School of Medicine, CA, USA
| | | | | |
Collapse
|
18
|
Lam P, Khan G, Stripecke R, Hui KM, Kasahara N, Peng KW, Guinn BA. The innovative evolution of cancer gene and cellular therapies. Cancer Gene Ther 2013; 20:141-9. [PMID: 23370333 DOI: 10.1038/cgt.2012.93] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We provide an overview of the latest developments in cancer gene therapy--from the bench to early-stage clinical trials. We describe the most recent work of worldwide teams including experienced scientists and clinicians, reflecting the recent emergence of gene therapy from the 'Valley of Death'. The treatment efficacy of clinical gene therapy has now been shown in a number of diseases including cancer and we are observing a renewed interest by big pharmaceutical and biotechnology companies most obviously demonstrated by Amgen's acquisition of Biovex for up to USD$1 billion. There is an opportunity to be cautiously hopeful regarding the future of gene therapy in the clinic and we review here some of the most recent progress in the field.
Collapse
Affiliation(s)
- P Lam
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
19
|
Amore A, Wals K, Koekoek E, Hoppes R, Toebes M, Schumacher TNM, Rodenko B, Ovaa H. Development of a hypersensitive periodate-cleavable amino acid that is methionine- and disulfide-compatible and its application in MHC exchange reagents for T cell characterisation. Chembiochem 2013; 14:123-31. [PMID: 23280887 PMCID: PMC3561698 DOI: 10.1002/cbic.201200540] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Indexed: 12/02/2022]
Abstract
Incorporation of cleavable linkers into peptides and proteins is of particular value in the study of biological processes. Here we describe the synthesis of a cleavable linker that is hypersensitive to oxidative cleavage as the result of the periodate reactivity of a vicinal amino alcohol moiety. Two strategies directed towards the synthesis of a building block suitable for solid-phase peptide synthesis were developed: a chemoenzymatic route, involving L-threonine aldolase, and an enantioselective chemical route; these led to α,γ-diamino-β-hydroxybutanoic acids in diastereoisomerically mixed and enantiopure forms, respectively. Incorporation of the 1,2-amino alcohol linker into the backbone of a peptide generated a conditional peptide that was rapidly cleaved at very low concentrations of sodium periodate. This cleavable peptide ligand was applied in the generation of MHC exchange reagents for the detection of antigen-specific T cells in peripheral blood cells. The extremely low concentration of periodate required to trigger MHC peptide exchange allowed the co-oxidation of methionine and disulfide residues to be avoided. Conditional MHC reagents hypersensitive to periodate can now be applied without limitations when UV irradiation is undesired or less practical.
Collapse
Affiliation(s)
- Alessia Amore
- Division of Cell Biology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands) E-mail:
| | - Kim Wals
- Division of Cell Biology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands) E-mail:
| | - Evelyn Koekoek
- Division of Cell Biology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands) E-mail:
| | - Rieuwert Hoppes
- Division of Cell Biology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands) E-mail:
| | - Mireille Toebes
- Division of Immunology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands)
| | - Ton N M Schumacher
- Division of Immunology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands)
| | - Boris Rodenko
- Division of Cell Biology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands) E-mail:
| | - Huib Ovaa
- Division of Cell Biology, The Netherlands Cancer InstitutePlesmanlaan 121, 1066 CX Amsterdam (the Netherlands) E-mail:
| |
Collapse
|
20
|
Underhill GH, Peter G, Chen CS, Bhatia SN. Bioengineering Methods for Analysis of Cells In Vitro. Annu Rev Cell Dev Biol 2012; 28:385-410. [DOI: 10.1146/annurev-cellbio-101011-155709] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Galie Peter
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Christopher S. Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Sangeeta N. Bhatia
- Division of Health Sciences and Technology,
- Department of Electrical Engineering and Computer Science,
- The Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
- Division of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115
| |
Collapse
|
21
|
Suchard MS. Missing: a diagnostic technique to enumerate antigen-specific T cells. Crit Rev Oncol Hematol 2012; 83:276-82. [PMID: 22137827 PMCID: PMC3496851 DOI: 10.1016/j.critrevonc.2011.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 10/26/2011] [Accepted: 11/09/2011] [Indexed: 12/14/2022] Open
Abstract
T lymphocytes are responsible for immune responses against pathogens, immune surveillance against cancer and maintenance of tolerance to self. While techniques available to detect antigen-specific T cells have been well described, there is a missing technique in our repertoire. While fluorescent multimers can be used for limited research applications, there is no existing technique suitable for detection of antigen-specific T cells in a diagnostic setting. The absence of such a technology has inhibited the search for "correlates of protection" against infectious, autoimmune or malignant disease. This critical review of existing methods will highlight the limitations of the data on which our current understanding of the immune system is based, in an effort to stimulate development of improved techniques.
Collapse
Affiliation(s)
- Melinda Shelley Suchard
- Molecular Medicine and Haematology, National Health Laboratory Service, Faculty of Health Sciences, University of Witwatersrand, 7 York Road Parktown, Johannesburg 2192, South Africa.
| |
Collapse
|
22
|
Papp K, Szittner Z, Prechl J. Life on a microarray: assessing live cell functions in a microarray format. Cell Mol Life Sci 2012; 69:2717-25. [PMID: 22391673 PMCID: PMC11115177 DOI: 10.1007/s00018-012-0947-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 02/14/2012] [Accepted: 02/16/2012] [Indexed: 01/07/2023]
Abstract
Microarray technology outgrew the detection of simple intermolecular interactions, as incubation of slides with living cells opened new vistas. Cell-based array technology permits simultaneous detection of several different cell surface molecules, allowing the complex characterization of cells with an amount of information that is hardly assessed by any other technique. Furthermore, binding of cells to printed antibodies or ligands may induce their activation, and consequently the outcome of these interactions, such as phosphorylation, gene expression, secretion of various products; differentiation, proliferation and apoptosis of the cells are also measurable on arrays. Moreover, since cells can be transfected with printed vectors, over- or under-expression of selected genes is also achievable simultaneously, creating a nice tool for assessing the function of a given gene. The enormously high-throughput cell-based microarray technology enables testing the effect of external stimuli on a scale that was earlier unthinkable. This review summarizes the possible applications of cell-based arrays.
Collapse
Affiliation(s)
- Krisztián Papp
- Immunology Research Group, Hungarian Academy of Sciences, MTA-ELTE, Pázmány P.s. 1/C, Budapest 1117, Hungary.
| | | | | |
Collapse
|
23
|
Revzin A, Maverakis E, Chang HC. Biosensors for immune cell analysis-A perspective. BIOMICROFLUIDICS 2012; 6:21301-2130113. [PMID: 22655003 PMCID: PMC3360707 DOI: 10.1063/1.4706845] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 04/10/2012] [Indexed: 05/14/2023]
Abstract
Massively parallel analysis of single immune cells or small immune cell colonies for disease detection, drug screening, and antibody production represents a "killer app" for the rapidly maturing microfabrication and microfluidic technologies. In our view, microfabricated solid-phase and flow cytometry platforms of the future will be complete with biosensors and electrical/mechanical/optical actuators and will enable multi-parametric analysis of cell function, real-time detection of secreted signals, and facile retrieval of cells deemed interesting.
Collapse
|
24
|
Sharivkin R, Walker MD, Soen Y. Proteomics-based dissection of human endoderm progenitors by differential cell capture on antibody array. Mol Cell Proteomics 2012; 11:586-95. [PMID: 22580589 DOI: 10.1074/mcp.m111.016840] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heterogeneity, shortage of material, and lack of progenitor-specific cell surface markers are major obstacles to elucidating the mechanisms underlying developmental processes. Here we report a proteomics platform that alleviates these difficulties and demonstrate its effectiveness in fractionating heterogeneous cultures of early endoderm derived from human embryonic stem cells. The approach, designated differential cell-capture antibody array, is based on highly parallel, comparative screening of live cell populations using hundreds of antibodies directed against cell-surface antigens. We used this platform to fractionate the hitherto unresolved early endoderm compartment of CXCR4+ cells and identify several endoderm (CD61+ and CD63+) and non-endoderm (CD271+, CD49F+, CD44+ and B2M+) sub-populations. We provide evidence that one of these sub-populations, CD61+, is directly derived from CXCR4+ cells, displays characteristic kinetics of emergence, and exhibits a distinct gene expression profile. The results demonstrate the potential of the cell-capture antibody array as a powerful proteomics tool for detailed dissection of heterogeneous cellular systems.
Collapse
Affiliation(s)
- Revital Sharivkin
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | | |
Collapse
|
25
|
Andersen RS, Kvistborg P, Frøsig TM, Pedersen NW, Lyngaa R, Bakker AH, Shu CJ, Straten PT, Schumacher TN, Hadrup SR. Parallel detection of antigen-specific T cell responses by combinatorial encoding of MHC multimers. Nat Protoc 2012; 7:891-902. [PMID: 22498709 DOI: 10.1038/nprot.2012.037] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fluorescently labeled multimeric complexes of peptide-MHC, the molecular entities recognized by the T cell receptor, have become essential reagents for detection of antigen-specific CD8(+) T cells by flow cytometry. Here we present a method for high-throughput parallel detection of antigen-specific T cells by combinatorial encoding of MHC multimers. Peptide-MHC complexes are produced by UV-mediated MHC peptide exchange and multimerized in the form of streptavidin-fluorochrome conjugates. Eight different fluorochromes are used for the generation of MHC multimers and, by a two-dimensional combinatorial matrix, these eight fluorochromes are combined to generate 28 unique two-color codes. By the use of combinatorial encoding, a large number of different T cell populations can be detected in a single sample. The method can be used for T cell epitope mapping, and also for the monitoring of CD8(+) immune responses during cancer and infectious disease or after immunotherapy. One panel of 28 combinatorially encoded MHC multimers can be prepared in 4 h. Staining and detection takes a further 3 h.
Collapse
Affiliation(s)
- Rikke Sick Andersen
- Center for Cancer Immune Therapy, Department of Hematology, University Hospital Herlev, Denmark
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Weiss VL, Lee TH, Song H, Kouo TS, Black CM, Sgouros G, Jaffee EM, Armstrong TD. Trafficking of high avidity HER-2/neu-specific T cells into HER-2/neu-expressing tumors after depletion of effector/memory-like regulatory T cells. PLoS One 2012; 7:e31962. [PMID: 22359647 PMCID: PMC3281086 DOI: 10.1371/journal.pone.0031962] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 01/16/2012] [Indexed: 02/02/2023] Open
Abstract
Background Cancer vaccines are designed to activate and enhance cancer-antigen-targeted T cells that are suppressed through multiple mechanisms of immune tolerance in cancer-bearing hosts. T regulatory cell (Treg) suppression of tumor-specific T cells is one barrier to effective immunization. A second mechanism is the deletion of high avidity tumor-specific T cells, which leaves a less effective low avidity tumor specific T cell repertoire available for activation by vaccines. Treg depleting agents including low dose cyclophosphamide (Cy) and antibodies that deplete CD25-expressing Tregs have been used with limited success to enhance the potency of tumor-specific vaccines. In addition, few studies have evaluated mechanisms that activate low avidity cancer antigen-specific T cells. Therefore, we developed high and low avidity HER-2/neu-specific TCR transgenic mouse colonies specific for the same HER-2/neu epitope to define the tolerance mechanisms that specifically affect high versus low avidity tumor-specific T cells. Methodology/Principal Findings High and low avidity CD8+ T cell receptor (TCR) transgenic mice specific for the breast cancer antigen HER-2/neu (neu) were developed to provide a purified source of naïve, tumor-specific T cells that can be used to study tolerance mechanisms. Adoptive transfer studies into tolerant FVB/N-derived HER-2/neu transgenic (neu-N) mice demonstrated that high avidity, but not low avidity, neu-specific T cells are inhibited by Tregs as the dominant tolerizing mechanism. High avidity T cells persisted, produced IFNγ, trafficked into tumors, and lysed tumors after adoptive transfer into mice treated with a neu-specific vaccine and low dose Cy to deplete Tregs. Analysis of Treg subsets revealed a Cy-sensitive CD4+Foxp3+CD25low tumor-seeking migratory phenotype, characteristic of effector/memory Tregs, and capable of high avidity T cell suppression. Conclusion/Significance Depletion of CD25low Tregs allows activation of tumor-clearing high avidity T cells. Thus, the development of agents that specifically deplete Treg subsets should translate into more effective immunotherapies while avoiding autoimmunity.
Collapse
Affiliation(s)
- Vivian L. Weiss
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Immunology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Timothy H. Lee
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Hong Song
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Radiology and Nuclear Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Theodore S. Kouo
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Immunology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chelsea M. Black
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Cellular and Molecular Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - George Sgouros
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Radiology and Nuclear Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Elizabeth M. Jaffee
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Immunology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Cellular and Molecular Medicine, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Graduate Program in Pharmacology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- The Skip Viragh Pancreatic Cancer Center, and Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Todd D. Armstrong
- The Sidney Kimmel Cancer Center at Johns Hopkins, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, John Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
28
|
Li Pira G, Ivaldi F, Manca F. Selective binding of CD4 and CD8 T-cells to antigen presenting cells for enrichment of CMV and HIV specific T-lymphocytes. J Immunol Methods 2012; 376:125-31. [DOI: 10.1016/j.jim.2012.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 12/29/2011] [Accepted: 01/03/2012] [Indexed: 10/14/2022]
|
29
|
Chen L, Liu X, Su B, Li J, Jiang L, Han D, Wang S. Aptamer-mediated efficient capture and release of T lymphocytes on nanostructured surfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:4376-4380. [PMID: 21882263 DOI: 10.1002/adma.201102435] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Revised: 07/15/2011] [Indexed: 05/31/2023]
Affiliation(s)
- Li Chen
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P. R. China
| | | | | | | | | | | | | |
Collapse
|
30
|
Davis MM, Altman JD, Newell EW. Interrogating the repertoire: broadening the scope of peptide-MHC multimer analysis. Nat Rev Immunol 2011; 11:551-8. [PMID: 21760610 PMCID: PMC3699324 DOI: 10.1038/nri3020] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Labelling antigen-specific T cells with peptide-MHC multimers has provided an invaluable way to monitor T cell-mediated immune responses. A number of recent developments in this technology have made these multimers much easier to make and use in large numbers. Furthermore, enrichment techniques have provided a greatly increased sensitivity that allows the analysis of the naive T cell repertoire directly. Thus, we can expect a flood of new information to emerge in the coming years.
Collapse
Affiliation(s)
- Mark M Davis
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA.
| | | | | |
Collapse
|
31
|
Cortès S, Villiers CL, Colpo P, Couderc R, Brakha C, Rossi F, Marche PN, Villiers MB. Biosensor for direct cell detection, quantification and analysis. Biosens Bioelectron 2011; 26:4162-8. [DOI: 10.1016/j.bios.2011.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 04/07/2011] [Accepted: 04/08/2011] [Indexed: 10/18/2022]
|
32
|
Rodríguez-Seguí SA, Pons Ximénez JI, Sevilla L, Ruiz A, Colpo P, Rossi F, Martínez E, Samitier J. Quantification of protein immobilization on substrates for cellular microarray applications. J Biomed Mater Res A 2011; 98:245-56. [PMID: 21626656 DOI: 10.1002/jbm.a.33089] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 10/11/2010] [Accepted: 02/03/2011] [Indexed: 11/07/2022]
Abstract
Cellular microarray developments and its applications are the next step after DNA and protein microarrays. The choice of the surface chemistry of the substrates used for the implementation of this technique, that must favor proper protein immobilization while avoiding cell adhesion on the nonspotted areas, presents a complex challenge. This is a key issue since usually the best nonfouling surfaces are also the ones that retain immobilized the smallest amounts of printed protein. To quantitatively assess the amount of protein immobilization, in this study several combinations of fluorescently labeled fibronectin (Fn*) and streptavidin (SA*) were microspotted, with and without glycerol addition in the printing buffer, on several substrates suitable for cellular microarrays. The substrates assayed included chemically activated surfaces as well as Poly ethylene oxide (PEO) films that are nonfouling in solution but accept adhesion of proteins in dry conditions. The results showed that the spotted Fn* was retained by all the surfaces, although the PEO surface did show smaller amounts of immobilization. The SA*, on the other hand, was only retained by the chemically activated surfaces. The inclusion of glycerol in the printing buffer significantly reduced the immobilization of both proteins. The results presented in this article provide quantitative evidence of the convenience of using a chemically activated surface to immobilize proteins relevant for cellular microarray applications, particularly when ECM proteins are cospotted with smaller factors which are more difficult to be retained by the surfaces.
Collapse
Affiliation(s)
- Santiago A Rodríguez-Seguí
- Nanobioengineering group, Institute for Bioengineering of Catalonia, Baldiri i Reixac 10-12, 08028 Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Adamczyk-Poplawska M, Markowicz S, Jagusztyn-Krynicka EK. Proteomics for development of vaccine. J Proteomics 2011; 74:2596-616. [PMID: 21310271 DOI: 10.1016/j.jprot.2011.01.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 01/26/2011] [Accepted: 01/31/2011] [Indexed: 12/20/2022]
Abstract
The success of genome projects has provided us with a vast amount of information on genes of many pathogenic species and has raised hopes for rapid progress in combating infectious diseases, both by construction of new effective vaccines and by creating a new generation of therapeutic drugs. Proteomics, a strategy complementary to the genomic-based approach, when combined with immunomics (looking for immunogenic proteins) and vaccinomics (characterization of host response to immunization), delivers valuable information on pathogen-host cell interaction. It also speeds the identification and detailed characterization of new antigens, which are potential candidates for vaccine development. This review begins with an overview of the global status of vaccinology based on WHO data. The main part of this review describes the impact of proteomic strategies on advancements in constructing effective antibacterial, antiviral and anticancer vaccines. Diverse aspects of disease mechanisms and disease preventions have been investigated by proteomics.
Collapse
Affiliation(s)
- Monika Adamczyk-Poplawska
- Department of Virology, Institute of Microbiology, Biology Faculty, Warsaw University, Warsaw, Poland
| | | | | |
Collapse
|
34
|
Prasad S, Cody V, Saucier-Sawyer JK, Saltzman WM, Sasaki CT, Edelson RL, Birchall MA, Hanlon DJ. Polymer nanoparticles containing tumor lysates as antigen delivery vehicles for dendritic cell-based antitumor immunotherapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2011; 7:1-10. [PMID: 20692374 PMCID: PMC3073408 DOI: 10.1016/j.nano.2010.07.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 07/19/2010] [Accepted: 07/19/2010] [Indexed: 01/17/2023]
Abstract
Encapsulation of tumor-associated antigens in polymer nanoparticles (NP) is a promising approach to enhance efficiency of antigen delivery for anti-tumor vaccines. Head and neck squamous carcinoma (HNSCC) cell lines were initially used to generate tumor-associated antigens (TAA)-containing poly (lactic-co-glycolic acid) (PLGA) NP; encapsulation efficiency and release kinetics were profiled. Findings were adopted to entrap fresh tumor lysate from five patients with advanced HNSCC. To test the hypothesis that NP enhance antigen presentation, dendritic cell (DC) produced from patient blood monocyte precursors were loaded with either the un-encapsulated or NP-encapsulated versions of tumor lysates. These were used to stimulate freshly-isolated autologous CD8+ T cells. In four of five patients, anti-tumor CD8+ T cells showed significantly increased immunostimulatory IFN-γ (p=0.071) or decreased immmunoinhibitory IL-10 production (p=0.0004) associated with NP-mediated antigen delivery. The observations represent an enabling step in the production of clinically-translatable, inexpensive, highly-efficient, and personalized polymer-based immunotherapy for solid organ malignancies. FROM THE CLINICAL EDITOR Enhancing the antigen presentation may be a viable approach to increase the efficiency of tumor cell directed cytotoxicity via immune mechanisms. This study presents an example for this using head and neck cancer cell lines and nanotechnology-based encapsulated antigen presentation to dendritic cells. The observed CD8+ T-cell response was significantly enhanced. This method may pave the way to a highly efficient cancer cell elimination method with minimal to no toxicity.
Collapse
Affiliation(s)
- Shashi Prasad
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| | - Virginia Cody
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| | | | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06520-8260, USA
| | - Clarence T. Sasaki
- Section of Otolaryngology, Yale University, New Haven, Connecticut 06520-8260, USA
| | - Richard L. Edelson
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| | | | - Douglas J. Hanlon
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| |
Collapse
|
35
|
Yue C, Oelke M, Paulaitis ME, Schneck JP. Novel cellular microarray assay for profiling T-cell peptide antigen specificities. J Proteome Res 2010; 9:5629-37. [PMID: 20836567 DOI: 10.1021/pr100447b] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We present a novel cellular microarray assay using soluble peptide-loaded HLA A2-Ig dimer complexes that optimizes the avidity of peptide-HLA binding by preserving the molecular flexibility of the dimer complex while attaining much higher concentrations of the complex relative to cognate T-cell receptors. A seminal advance in assay development is made by separating the molecular T-cell receptor recognition event from the binding interactions that lead to antigen-specific cell capture on the microarray. This advance enables the quantitative determination of antigen-specific frequencies in heterogeneous T-cell populations without enumerating the number of cells captured on the microarray. The specificity of cell capture, sensitivity to low antigen-specific frequencies, and quantitation of antigenic T-cell specificities are established using CD8 T-cell populations with prepared antigen-specific CTL frequencies and heterogeneous T cells isolated from peripheral blood. The results demonstrate several advantages for high-throughput broad-based, quantitative assessments of low-frequency antigen specificities. The assay enables the use of cellular microarrays to determine the stability and flux of antigen-specific T-cell responses within and across populations.
Collapse
Affiliation(s)
- C Yue
- Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
36
|
Hoff A, Bagû AC, André T, Roth G, Wiesmüller KH, Gückel B, Brock R. Peptide microarrays for the profiling of cytotoxic T-lymphocyte activity using minimum numbers of cells. Cancer Immunol Immunother 2010; 59:1379-87. [PMID: 20512327 PMCID: PMC2892610 DOI: 10.1007/s00262-010-0867-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 04/30/2010] [Indexed: 11/30/2022]
Abstract
The identification of epitopes that elicit cytotoxic T-lymphocyte activity is a prerequisite for the development of cancer-specific immunotherapies. However, especially the parallel characterization of several epitopes is limited by the availability of T cells. Microarrays have enabled an unprecedented miniaturization and parallelization in biological assays. Here, we developed peptide microarrays for the detection of CTL activity. MHC class I-binding peptide epitopes were pipetted onto polymer-coated glass slides. Target cells, loaded with the cell-impermeant dye calcein, were incubated on these arrays, followed by incubation with antigen-expanded CTLs. Cytotoxic activity was detected by release of calcein and detachment of target cells. With only 200,000 cells per microarray, CTLs could be detected at a frequency of 0.5% corresponding to 1,000 antigen-specific T cells. Target cells and CTLs only settled on peptide spots enabling a clear separation of individual epitopes. Even though no physical boundaries were present between the individual spots, peptide loading only occurred locally and cytolytic activity was confined to the spots carrying the specific epitope. The peptide microarrays provide a robust platform that implements the whole process from antigen presentation to the detection of CTL activity in a miniaturized format. The method surpasses all established methods in the minimum numbers of cells required. With antigen uptake occurring on the microarray, further applications are foreseen in the testing of antigen precursors that require uptake and processing prior to presentation.
Collapse
Affiliation(s)
- Antje Hoff
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- Present Address: Trinity Centre for Health Sciences, Institute for Molecular Medicine, Trinity College Dublin, St. James Street, Dublin 8, Ireland
| | - Ana-Cristina Bagû
- Department of Gynecology and Obstetrics, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Thomas André
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- Present Address: Bachem AG, Hauptstrasse 144, 4416 Bubendorf, Switzerland
| | - Günter Roth
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Georges-Koehler-Allee 106, 79110 Freiburg, Germany
| | | | - Brigitte Gückel
- Department of Gynecology and Obstetrics, University Hospital Tübingen, Calwerstraße 7, 72076 Tübingen, Germany
| | - Roland Brock
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
37
|
Hadrup SR, Schumacher TN. MHC-based detection of antigen-specific CD8+ T cell responses. Cancer Immunol Immunother 2010; 59:1425-33. [PMID: 20177676 PMCID: PMC2892606 DOI: 10.1007/s00262-010-0824-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 01/30/2010] [Indexed: 12/05/2022]
Abstract
The hallmark of adaptive immunity is its ability to recognise a wide range of antigens and technologies that capture this diversity are therefore of substantial interest. New methods have recently been developed that allow the parallel analysis of T cell reactivity against vast numbers of different epitopes in limited biological material. These technologies are based on the joint binding of differentially labelled MHC multimers on the T cell surface, thereby providing each antigen-specific T cell population with a unique multicolour code. This strategy of 'combinatorial encoding' enables detection of many (at least 25) different T cell populations per sample and should be of broad value for both T cell epitope identification and immunomonitoring.
Collapse
Affiliation(s)
- Sine Reker Hadrup
- Department of Hematology, Center for Cancer Immune Therapy, CCIT, University Hospital Herlev, Herlev, Denmark.
| | | |
Collapse
|
38
|
Hoppes R, Ekkebus R, Schumacher TN, Ovaa H. Technologies for MHC class I immunoproteomics. J Proteomics 2010; 73:1945-53. [DOI: 10.1016/j.jprot.2010.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 05/06/2010] [Accepted: 05/16/2010] [Indexed: 11/29/2022]
|
39
|
Ge X, Gebe JA, Bollyky PL, James EA, Yang J, Stern LJ, Kwok WW. Peptide-MHC cellular microarray with innovative data analysis system for simultaneously detecting multiple CD4 T-cell responses. PLoS One 2010; 5:e11355. [PMID: 20634998 PMCID: PMC2902358 DOI: 10.1371/journal.pone.0011355] [Citation(s) in RCA: 16] [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: 03/26/2010] [Accepted: 06/07/2010] [Indexed: 11/21/2022] Open
Abstract
Background Peptide:MHC cellular microarrays have been proposed to simultaneously characterize multiple Ag-specific populations of T cells. The practice of studying immune responses to complicated pathogens with this tool demands extensive knowledge of T cell epitopes and the availability of peptide:MHC complexes for array fabrication as well as a specialized data analysis approach for result interpretation. Methodology/Principal Findings We co-immobilized peptide:DR0401 complexes, anti-CD28, anti-CD11a and cytokine capture antibodies on the surface of chamber slides to generate a functional array that was able to detect rare Ag-specific T cell populations from previously primed in vitro T cell cultures. A novel statistical methodology was also developed to facilitate batch processing of raw array-like data into standardized endpoint scores, which linearly correlated with total Ag-specific T cell inputs. Applying these methods to analyze Influenza A viral antigen-specific T cell responses, we not only revealed the most prominent viral epitopes, but also demonstrated the heterogeneity of anti-viral cellular responses in healthy individuals. Applying these methods to examine the insulin producing beta-cell autoantigen specific T cell responses, we observed little difference between autoimmune diabetic patients and healthy individuals, suggesting a more subtle association between diabetes status and peripheral autoreactive T cells. Conclusions/Significance The data analysis system is reliable for T cell specificity and functional testing. Peptide:MHC cellular microarrays can be used to obtain multi-parametric results using limited blood samples in a variety of translational settings.
Collapse
Affiliation(s)
- Xinhui Ge
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - John A. Gebe
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - Paul L. Bollyky
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - Eddie A. James
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - Junbao Yang
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
| | - Lawrence J. Stern
- Department of Pathology, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - William W. Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
40
|
High throughput T epitope mapping and vaccine development. J Biomed Biotechnol 2010; 2010:325720. [PMID: 20617148 PMCID: PMC2896667 DOI: 10.1155/2010/325720] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 02/18/2010] [Accepted: 04/20/2010] [Indexed: 11/22/2022] Open
Abstract
Mapping of antigenic peptide sequences from proteins of relevant pathogens recognized by T helper (Th) and by cytolytic T lymphocytes (CTL) is crucial for vaccine development. In fact, mapping of T-cell epitopes provides useful information for the design of peptide-based vaccines and of peptide libraries to monitor specific cellular immunity in protected individuals, patients and vaccinees. Nevertheless, epitope mapping is a challenging task. In fact, large panels of overlapping peptides need to be tested with lymphocytes to identify the sequences that induce a T-cell response. Since numerous peptide panels from antigenic proteins are to be screened, lymphocytes available from human subjects are a limiting factor. To overcome this limitation, high throughput (HTP) approaches based on miniaturization and automation of T-cell assays are needed. Here we consider the most recent applications of the HTP approach to T epitope mapping. The alternative or complementary use of in silico prediction and experimental epitope definition is discussed in the context of the recent literature. The currently used methods are described with special reference to the possibility of applying the HTP concept to make epitope mapping an easier procedure in terms of time, workload, reagents, cells and overall cost.
Collapse
|
41
|
Abstract
CD8(+) T cells (also called cytotoxic T lymphocytes) play a major role in protective immunity against many infectious pathogens and can eradicate malignant cells. The path from naive precursor to effector and memory CD8(+) T-cell development begins with interactions between matured antigen-bearing dendritic cells (DCs) and antigen-specific naive T-cell clonal precursors. By integrating differences in antigenic, costimulatory, and inflammatory signals, a developmental program is established that governs many key parameters associated with the ensuing response, including the extent and magnitude of clonal expansion, the functional capacities of the effector cells, and the size of the memory pool that survives after the contraction phase. In this review, we discuss the multitude of signals that drive effector and memory CD8(+) T-cell differentiation and how the differences in the nature of these signals contribute to the diversity of CD8(+) T-cell responses.
Collapse
Affiliation(s)
- Ramon Arens
- Laboratory of Cellular Immunology, The La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Stephen P. Schoenberger
- Laboratory of Cellular Immunology, The La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| |
Collapse
|
42
|
Analysis of vaccine-induced T cells in humans with cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:178-88. [PMID: 20795549 DOI: 10.1007/978-1-4419-6451-9_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Over the past several years, progress in the field of tumor immunology has lead to advances in active immunotherapy and vaccination as a means ofeliciting tumor-specific immune responses to mediate tumor regression and clearance. Developing vaccines targeted against cancer became an important focus as a therapy following the success of viral vaccines in preventing infection and disease. In humans with cancer, similar to viral infections, the host immune system is capable of recognizing antigens expressed on tumor cells. This similarity allows the immunological framework of the viral vaccine to be adapted to the cancer setting in hopes of enhancing human T-cell reactivity against tumor. It is generally believed that a requirement for tumor destruction to occur is the induction of sufficient levels of immune cells with high avidity for recognition of tumor antigens. Moreover, the cells must be targeted to the tumor site and be capable of infiltrating tumor stroma.2 Several tumor-associated antigens (TAA) have been identified in the melanoma model which has allowed for immunization trials to evaluate therapeutic potential of tumor-specific T-cell induction. Some clinical trials reported limited success ofT-cell mediated tumor rejection, reportingpartial or complete regression in 10 to 30% of patients. Although tumor regression was not observed following active immunization in vivo, ex vivo assays evaluating TAA-specific T cells demonstrated tumor recognition and subsequent T-cell activation suggesting that tumor-specific T-cell induction indeed occurs but alone is not adequate to induce tumor regression. Recently, the usefulness and success of active-specific immunization (ASI) against TAAs as a means ofeliciting a tumor-specific immune response leading to tumor regression and clearance has been a topic of debate and discussion.
Collapse
|
43
|
Rodenko B, Toebes M, Celie PHN, Perrakis A, Schumacher TNM, Ovaa H. Class I major histocompatibility complexes loaded by a periodate trigger. J Am Chem Soc 2009; 131:12305-13. [PMID: 19655751 DOI: 10.1021/ja9037565] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Class I major histocompatibility complexes (MHCs) present peptide ligands on the cell surface for recognition by appropriate cytotoxic T cells. The unstable nature of unliganded MHC necessitates the production of recombinant class I complexes through in vitro refolding reactions in the presence of an added excess of peptides. This strategy is not amenable to high-throughput production of vast collections of class I complexes. To address this issue, we recently designed photocaged MHC ligands that can be cleaved by a UV light trigger in the MHC bound state under conditions that do not affect the integrity of the MHC structure. The results obtained with photocaged MHC ligands demonstrate that conditional MHC ligands can form a generally applicable concept for the creation of defined peptide-MHCs. However, the use of UV exposure to mediate ligand exchange is unsuited for a number of applications, due to the lack of UV penetration through cell culture systems and due to the transfer of heat upon UV irradiation, which can induce evaporation. To overcome these limitations, here, we provide proof-of-concept for the generation of defined peptide-MHCs by chemical trigger-induced ligand exchange. The crystal structure of the MHC with the novel chemosensitive ligand showcases that the ligand occupies the expected binding site, in a conformation where the hydroxyl groups should be reactive to periodate. We proceed to validate this technology by producing peptide-MHCs that can be used for T cell detection. The methodology that we describe here should allow loading of MHCs with defined peptides in cell culture devices, thereby permitting antigen-specific T cell expansion and purification for cell therapy. In addition, this technology will be useful to develop miniaturized assay systems for performing high-throughput screens for natural and unnatural MHC ligands.
Collapse
Affiliation(s)
- Boris Rodenko
- Division of Cell Biology II, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
44
|
Maynard JA, Lindquist NC, Sutherland JN, Lesuffleur A, Warrington AE, Rodriguez M, Oh SH. Surface plasmon resonance for high-throughput ligand screening of membrane-bound proteins. Biotechnol J 2009; 4:1542-58. [PMID: 19918786 PMCID: PMC2790208 DOI: 10.1002/biot.200900195] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Technologies based on surface plasmon resonance (SPR) have allowed rapid, label-free characterization of protein-protein and protein-small molecule interactions. SPR has become the gold standard in industrial and academic settings, in which the interaction between a pair of soluble binding partners is characterized in detail or a library of molecules is screened for binding against a single soluble protein. In spite of these successes, SPR is only beginning to be adapted to the needs of membrane-bound proteins which are difficult to study in situ but represent promising targets for drug and biomarker development. Existing technologies, such as BIAcoreTM, have been adapted for membrane protein analysis by building supported lipid layers or capturing lipid vesicles on existing chips. Newer technologies, still in development, will allow membrane proteins to be presented in native or near-native formats. These include SPR nanopore arrays, in which lipid bilayers containing membrane proteins stably span small pores that are addressable from both sides of the bilayer. Here, we discuss current SPR instrumentation and the potential for SPR nanopore arrays to enable quantitative, high-throughput screening of G protein coupled receptor ligands and applications in basic cellular biology.
Collapse
Affiliation(s)
- Jennifer A. Maynard
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, 78705
| | - Nathan C. Lindquist
- Department of Electrical and Computer Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| | - Jamie N. Sutherland
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, 78705
| | - Antoine Lesuffleur
- Department of Electrical and Computer Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| | | | - Moses Rodriguez
- Departments of Neurology, Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455
| |
Collapse
|
45
|
Oble DA, Loewe R, Yu P, Mihm MC. Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human melanoma. CANCER IMMUNITY 2009; 9:3. [PMID: 19338264 PMCID: PMC2935762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Tumors contain variable numbers of lymphocytes, referred to as tumor infiltrating lymphocytes (TILs). In melanoma, the intensity of this lymphocytic infiltrate is believed to correlate with outcome, though there is some debate about the applicability of this finding for all melanomas. Much research has gone into classifying TILs with respect to antigen receptor structure and the antigen to which melanoma-specific T cells react. However, these studies for the most part did not immunophenotype TILs, and recent data has revealed that the composition of tumoral lymphocytes is not homogenous, but rather represents varying contributions from many lymphocytic subsets. Furthermore, the function of TILs is often compromised as a result of the accumulation of immunoregulatory cells and various tumor escape mechanisms. These recent insights stress the need to collect more data on the composition and function of TIL infiltrates before definitive conclusions about the prognostic significance of TILs can be drawn. Advances in immunology have also facilitated the development of immunotherapeutic strategies, examples of which will be discussed with a special emphasis on blocking antibodies against CTLA-4, which are prototypical immunotherapeutic agents. This flurry of novel "biological" therapies will undoubtedly complicate our already incomplete understanding of TIL immunobiology as each of these agents has the potential to uniquely distort the series of immunological events which normally occur in untreated melanoma. Therefore, considerable research is needed to better elucidate the function and prognostic significance of TILs in both untreated melanoma and tumors treated with "biological" therapy.
Collapse
MESH Headings
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD/immunology
- Antigens, CD/metabolism
- CTLA-4 Antigen
- Cytokines/immunology
- Cytokines/metabolism
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Immunologic Factors/immunology
- Immunologic Factors/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Melanoma/drug therapy
- Melanoma/immunology
- Melanoma/metabolism
- Prognosis
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Darryl A. Oble
- Department of Pathology, Massachusetts
General Hospital, Harvard Medical School55
Fruit Street, Warren 827Boston, MA 02114USA
- Department of Medicine, Section of Dermatology,
University of Chicago5841 S. Maryland Avenue, MC
5067Chicago, IL 60637USA
| | - Robert Loewe
- Department of Pathology, Massachusetts
General Hospital, Harvard Medical School55
Fruit Street, Warren 827Boston, MA 02114USA
| | - Ping Yu
- Department of Medicine, Section of Dermatology,
University of Chicago5841 S. Maryland Avenue, MC
5067Chicago, IL 60637USA
| | - Martin C. Mihm
- Department of Pathology, Massachusetts
General Hospital, Harvard Medical School55
Fruit Street, Warren 827Boston, MA 02114USA
- Department of Dermatology, Massachusetts
General Hospital, Harvard Medical School50
Staniford Street, Suite 200Boston, MA 02114USA
| |
Collapse
|
46
|
Shiina T, Hosomichi K, Inoko H, Kulski JK. The HLA genomic loci map: expression, interaction, diversity and disease. J Hum Genet 2009; 54:15-39. [PMID: 19158813 DOI: 10.1038/jhg.2008.5] [Citation(s) in RCA: 497] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The human leukocyte antigen (HLA) super-locus is a genomic region in the chromosomal position 6p21 that encodes the six classical transplantation HLA genes and at least 132 protein coding genes that have important roles in the regulation of the immune system as well as some other fundamental molecular and cellular processes. This small segment of the human genome has been associated with more than 100 different diseases, including common diseases, such as diabetes, rheumatoid arthritis, psoriasis, asthma and various other autoimmune disorders. The first complete and continuous HLA 3.6 Mb genomic sequence was reported in 1999 with the annotation of 224 gene loci, including coding and non-coding genes that were reviewed extensively in 2004. In this review, we present (1) an updated list of all the HLA gene symbols, gene names, expression status, Online Mendelian Inheritance in Man (OMIM) numbers, including new genes, and latest changes to gene names and symbols, (2) a regional analysis of the extended class I, class I, class III, class II and extended class II subregions, (3) a summary of the interspersed repeats (retrotransposons and transposons), (4) examples of the sequence diversity between different HLA haplotypes, (5) intra- and extra-HLA gene interactions and (6) some of the HLA gene expression profiles and HLA genes associated with autoimmune and infectious diseases. Overall, the degrees and types of HLA super-locus coordinated gene expression profiles and gene variations have yet to be fully elucidated, integrated and defined for the processes involved with normal cellular and tissue physiology, inflammatory and immune responses, and autoimmune and infectious diseases.
Collapse
Affiliation(s)
- Takashi Shiina
- Division of Basic Medical Science and Molecular Medicine, Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Japan.
| | | | | | | |
Collapse
|
47
|
Abstract
Recombinant major histocompatibility complex (MHC) class I molecules complexed with pathogen-specific or other disease-associated antigens have become essential reagents for the analysis of adaptive T-cell responses. However, conventional techniques for the production of recombinant peptide-MHC (pMHC) complexes are highly involved and thereby limit the use of pMHC complexes in terms of antigen diversity. To make pMHC-based techniques suitable for high-throughput analyses we developed an MHC peptide exchange technology based on the use of conditional MHC ligands. This technology enables the parallel production of thousands of different pMHC complexes within hours, allowing the development of high-throughput MHC-based assay systems to identify MHC ligands and cytotoxic T-cell responses. These high-throughput assays should prove valuable for the screening of entire disease-associated proteomes, including pathogen-encoded proteomes, tumor-associated antigens, and autoimmune antigens.
Collapse
|
48
|
Zhu H, Stybayeva G, Macal M, Ramanculov E, George MD, Dandekar S, Revzin A. A microdevice for multiplexed detection of T-cell-secreted cytokines. LAB ON A CHIP 2008; 8:2197-205. [PMID: 19023487 DOI: 10.1039/b810244a] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cytokines are produced by immune cells in response to viral or bacterial pathogens and therefore have significant diagnostic value. The goal of the present study was to develop a miniature device for detection of interleukin (IL)-2 and interferon (IFN)-gamma cytokines secreted by a small population of CD4 and CD8 T-cells. Microarrays of T-cell- and cytokine-specific Ab spots were printed onto poly(ethylene glycol) (PEG) hydrogel-coated glass slides and enclosed inside a microfluidic device, creating a miniature ( approximately 3 microL) immunoreaction chamber. Introduction of the red blood cell (RBC) depleted whole human blood into the microfluidic device followed by washing at a pre-defined shear stress resulted in isolation of pure CD4 and CD8 T-cells on their respective Ab spots. Importantly, the cells became localized next to anti-IL-2 and -IFN-gamma Ab spots. Mitogenic activation of the captured T-cells was followed by immunofluorescent staining (all steps carried out inside a microfluidic device), revealing concentration gradients of surface-bound cytokine molecules. A microarray scanner was then used to quantify the concentration of IFN-gamma and IL-2 near CD4 and CD8 T-cells. This study represents one of the first demonstrations of a microdevice for capturing desired T-cell subsets from a small blood volume and determining, on-chip, cytokine profiles of the isolated cells. Such a microdevice is envisioned as an immunology tool for multi-parametric analysis of T-cell function with direct applications in diagnosis/monitoring of HIV and other infectious diseases.
Collapse
Affiliation(s)
- He Zhu
- Department of Biomedical Engineering, University of California, Davis, 451 East Health Sciences St. #2619, Davis, CA 95616, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Huse M, Quann EJ, Davis MM. Shouts, whispers and the kiss of death: directional secretion in T cells. Nat Immunol 2008; 9:1105-11. [PMID: 18800163 PMCID: PMC2905669 DOI: 10.1038/ni.f.215] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
T cells use secreted soluble factors for highly specific intercellular communication and targeted cell killing. This specificity is achieved first through T cell receptor-mediated recognition of complexes of peptide and major histocompatibility complex displayed by appropriate antigen-presenting cells and then by the directed secretion of cytokines and lytic factors into the immunological synapse between the T cell and antigen-presenting cell. Studies have begun to probe the molecular basis for this synaptic secretion and have also shown that T cells release chemokines and certain inflammatory factors through a multidirectional pathway directed away from the synapse. Thus, the mode of secretion seems to be tailored to the intended function of the secreted molecule.
Collapse
Affiliation(s)
- Morgan Huse
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | | |
Collapse
|
50
|
Bakker AH, Hoppes R, Linnemann C, Toebes M, Rodenko B, Berkers CR, Hadrup SR, van Esch WJE, Heemskerk MHM, Ovaa H, Schumacher TNM. Conditional MHC class I ligands and peptide exchange technology for the human MHC gene products HLA-A1, -A3, -A11, and -B7. Proc Natl Acad Sci U S A 2008; 105:3825-30. [PMID: 18308940 PMCID: PMC2268811 DOI: 10.1073/pnas.0709717105] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Indexed: 01/28/2023] Open
Abstract
Major histocompatibility complex (MHC) class I multimer technology has become an indispensable immunological assay system to dissect antigen-specific cytotoxic CD8(+) T cell responses by flow cytometry. However, the development of high-throughput assay systems, in which T cell responses against a multitude of epitopes are analyzed, has been precluded by the fact that for each T cell epitope, a separate in vitro MHC refolding reaction is required. We have recently demonstrated that conditional ligands that disintegrate upon exposure to long-wavelength UV light can be designed for the human MHC molecule HLA-A2. To determine whether this peptide-exchange technology can be developed into a generally applicable approach for high throughput MHC based applications we set out to design conditional ligands for the human MHC gene products HLA-A1, -A3, -A11, and -B7. Here, we describe the development and characterization of conditional ligands for this set of human MHC molecules and apply the peptide-exchange technology to identify melanoma-associated peptides that bind to HLA-A3 with high affinity. The conditional ligand technology developed here will allow high-throughput MHC-based analysis of cytotoxic T cell immunity in the vast majority of Western European individuals.
Collapse
Affiliation(s)
| | - Rieuwert Hoppes
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | | | | - Boris Rodenko
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Celia R. Berkers
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | | | | - Mirjam H. M. Heemskerk
- Department of Hematology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Huib Ovaa
- Cellular Biochemistry, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | | |
Collapse
|