1
|
Binayke A, Zaheer A, Vishwakarma S, Singh S, Sharma P, Chandwaskar R, Gosain M, Raghavan S, Murugesan DR, Kshetrapal P, Thiruvengadam R, Bhatnagar S, Pandey AK, Garg PK, Awasthi A. A quest for universal anti-SARS-CoV-2 T cell assay: systematic review, meta-analysis, and experimental validation. NPJ Vaccines 2024; 9:3. [PMID: 38167915 PMCID: PMC10762233 DOI: 10.1038/s41541-023-00794-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Measuring SARS-CoV-2-specific T cell responses is crucial to understanding an individual's immunity to COVID-19. However, high inter- and intra-assay variability make it difficult to define T cells as a correlate of protection against COVID-19. To address this, we performed systematic review and meta-analysis of 495 datasets from 94 original articles evaluating SARS-CoV-2-specific T cell responses using three assays - Activation Induced Marker (AIM), Intracellular Cytokine Staining (ICS), and Enzyme-Linked Immunospot (ELISPOT), and defined each assay's quantitative range. We validated these ranges using samples from 193 SARS-CoV-2-exposed individuals. Although IFNγ ELISPOT was the preferred assay, our experimental validation suggested that it under-represented the SARS-CoV-2-specific T cell repertoire. Our data indicate that a combination of AIM and ICS or FluoroSpot assay would better represent the frequency, polyfunctionality, and compartmentalization of the antigen-specific T cell responses. Taken together, our results contribute to defining the ranges of antigen-specific T cell assays and propose a choice of assay that can be employed to better understand the cellular immune response against viral diseases.
Collapse
Affiliation(s)
- Akshay Binayke
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
- Jawaharlal Nehru University, New Delhi, India
| | - Aymaan Zaheer
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Siddhesh Vishwakarma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Savita Singh
- Translational Health Science and Technology Institute, Faridabad, India
| | - Priyanka Sharma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Rucha Chandwaskar
- Department of Microbiology, AMITY University Rajasthan, Jaipur, India
| | - Mudita Gosain
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | | | - Ramachandran Thiruvengadam
- Translational Health Science and Technology Institute, Faridabad, India
- Pondicherry Institute of Medical Sciences, Puducherry, India
| | | | | | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, India
- All India Institute of Medical Sciences, New Delhi, India
| | - Amit Awasthi
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India.
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India.
| |
Collapse
|
2
|
Arif S, Domingo-Vila C, Pollock E, Christakou E, Williams E, Tree TIM. Monitoring islet specific immune responses in type 1 diabetes clinical immunotherapy trials. Front Immunol 2023; 14:1183909. [PMID: 37283770 PMCID: PMC10240960 DOI: 10.3389/fimmu.2023.1183909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/02/2023] [Indexed: 06/08/2023] Open
Abstract
The number of immunotherapeutic clinical trials in type 1 diabetes currently being conducted is expanding, and thus there is a need for robust immune-monitoring assays which are capable of detecting and characterizing islet specific immune responses in peripheral blood. Islet- specific T cells can serve as biomarkers and as such can guide drug selection, dosing regimens and immunological efficacy. Furthermore, these biomarkers can be utilized in patient stratification which can then benchmark suitability for participation in future clinical trials. This review focusses on the commonly used immune-monitoring techniques including multimer and antigen induced marker assays and the potential to combine these with single cell transcriptional profiling which may provide a greater understanding of the mechanisms underlying immuno-intervention. Although challenges remain around some key areas such as the need for harmonizing assays, technological advances mean that multiparametric information derived from a single sample can be used in coordinated efforts to harmonize biomarker discovery and validation. Moreover, the technologies discussed here have the potential to provide a unique insight on the effect of therapies on key players in the pathogenesis of T1D that cannot be obtained using antigen agnostic approaches.
Collapse
|
3
|
Brudno JN, Lam N, Vanasse D, Shen YW, Rose JJ, Rossi J, Xue A, Bot A, Scholler N, Mikkilineni L, Roschewski M, Dean R, Cachau R, Youkharibache P, Patel R, Hansen B, Stroncek DF, Rosenberg SA, Gress RE, Kochenderfer JN. Safety and feasibility of anti-CD19 CAR T cells with fully human binding domains in patients with B-cell lymphoma. Nat Med 2020; 26:270-280. [PMID: 31959992 PMCID: PMC7781235 DOI: 10.1038/s41591-019-0737-3] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/10/2019] [Indexed: 12/30/2022]
Abstract
Anti-CD19 chimeric antigen receptor (CAR)-expressing T cells are an effective treatment for B-cell lymphoma, but often cause neurologic toxicity. We treated 20 patients with B-cell lymphoma on a phase I, first-in-human clinical trial of T cells expressing the new anti-CD19 CAR Hu19-CD828Z (NCT02659943). The primary objective was to assess safety and feasibility of Hu19-CD828Z T-cell therapy. Secondary objectives included assessments of blood levels of CAR T cells, anti-lymphoma activity, second infusions and immunogenicity. All objectives were met. Fifty-five percent of patients who received Hu19-CD828Z T cells obtained complete remission. Hu19-CD828Z T cells had clinical anti-lymphoma activity similar to that of T cells expressing FMC63-28Z, an anti-CD19 CAR tested previously by our group, which contains murine binding domains and is used in axicabtagene ciloleucel. However, severe neurologic toxicity occurred in only 5% of patients who received Hu19-CD828Z T cells, whereas 50% of patients who received FMC63-28Z T cells experienced this degree of toxicity (P = 0.0017). T cells expressing Hu19-CD828Z released lower levels of cytokines than T cells expressing FMC63-28Z. Lower levels of cytokines were detected in blood from patients who received Hu19-CD828Z T cells than in blood from those who received FMC63-28Z T cells, which could explain the lower level of neurologic toxicity associated with Hu19-CD828Z. Levels of cytokines released by CAR-expressing T cells particularly depended on the hinge and transmembrane domains included in the CAR design.
Collapse
Affiliation(s)
- Jennifer N Brudno
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Norris Lam
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Danielle Vanasse
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Jeremy J Rose
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | | | | | | | - Lekha Mikkilineni
- Medical Oncology/Hematology Fellowship Program, NCI, Bethesda, MD, USA
| | - Mark Roschewski
- Lymphoid Malignancy Branch, Center for Cancer Research, NCI, NIH Bethesda, Bethesda, MD, USA
| | - Robert Dean
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH, USA
| | - Raul Cachau
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Philippe Youkharibache
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rashmika Patel
- Office of Regulatory Affairs, NCI, NIH, Bethesda, MD, USA
| | - Brenna Hansen
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - David F Stroncek
- Center for Cellular Engineering, Clinical Center, NIH, Bethesda, MD, USA
| | - Steven A Rosenberg
- Surgery Branch, Center for Cancer Research, NCI, NIH Bethesda, Bethesda, MD, USA
| | - Ronald E Gress
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - James N Kochenderfer
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
4
|
Heide J, Vaughan KC, Sette A, Jacobs T, Schulze Zur Wiesch J. Comprehensive Review of Human Plasmodium falciparum-Specific CD8+ T Cell Epitopes. Front Immunol 2019; 10:397. [PMID: 30949162 PMCID: PMC6438266 DOI: 10.3389/fimmu.2019.00397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
Control of malaria is an important global health issue and there is still an urgent need for the development of an effective prophylactic vaccine. Multiple studies have provided strong evidence that Plasmodium falciparum-specific MHC class I-restricted CD8+ T cells are important for sterile protection against Plasmodium falciparum infection. Here, we present an interactive epitope map of all P. falciparum-specific CD8+ T cell epitopes published to date, based on a comprehensive data base (IEDB), and literature search. The majority of the described P. falciparum-specific CD8+ T cells were directed against the antigens CSP, TRAP, AMA1, and LSA1. Notably, most of the epitopes were discovered in vaccine trials conducted with malaria-naïve volunteers. Only few immunological studies of P. falciparum-specific CD8+ T cell epitopes detected in patients suffering from acute malaria or in people living in malaria endemic areas have been published. Further detailed immunological mappings of P. falciparum-specific epitopes of a broader range of P. falciparum proteins in different settings and with different disease status are needed to gain a more comprehensive understanding of the role of CD8+ T cell responses for protection, and to better guide vaccine design and to study their efficacy.
Collapse
Affiliation(s)
- Janna Heide
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Kerrie C Vaughan
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard-Nocht-Institute of Tropical Medicine, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
5
|
Barabas S, Spindler T, Kiener R, Tonar C, Lugner T, Batzilla J, Bendfeldt H, Rascle A, Asbach B, Wagner R, Deml L. An optimized IFN-γ ELISpot assay for the sensitive and standardized monitoring of CMV protein-reactive effector cells of cell-mediated immunity. BMC Immunol 2017; 18:14. [PMID: 28270111 PMCID: PMC5339961 DOI: 10.1186/s12865-017-0195-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/10/2017] [Indexed: 12/04/2022] Open
Abstract
Background In healthy individuals, Cytomegalovirus (CMV) infection is efficiently controlled by CMV-specific cell-mediated immunity (CMI). Functional impairment of CMI in immunocompromized individuals however can lead to uncontrolled CMV replication and severe clinical complications. Close monitoring of CMV-specific CMI is therefore clinically relevant and might allow a reliable prognosis of CMV disease as well as assist personalized therapeutic decisions. Methods Objective of this work was the optimization and technical validation of an IFN-γ ELISpot assay for a standardized, sensitive and reliable quantification of CMV-reactive effector cells. T-activated® immunodominant CMV IE-1 and pp65 proteins were used as stimulants. All basic assay parameters and reagents were tested and optimized to establish a user-friendly protocol and maximize the signal-to-noise ratio of the ELISpot assay. Results Optimized and standardized ELISpot revealed low intra-assay, inter-assay and inter-operator variability (coefficient of variation CV below 22%) and CV inter-site was lower than 40%. Good assay linearity was obtained between 6 × 104 and 2 × 105 PBMC per well upon stimulation with T-activated® IE-1 (R2 = 0.97) and pp65 (R2 = 0.99) antigens. Remarkably, stimulation of peripheral blood mononuclear cells (PBMC) with T-activated® IE-1 and pp65 proteins resulted in the activation of a broad range of CMV-reactive effector cells, including CD3+CD4+ (Th), CD3+CD8+ (CTL), CD3−CD56+ (NK) and CD3+CD56+ (NKT-like) cells. Accordingly, the optimized IFN-γ ELISpot assay revealed very high sensitivity (97%) in a cohort of 45 healthy donors, of which 32 were CMV IgG-seropositive. Conclusion The combined use of T-activated® IE-1 and pp65 proteins for the stimulation of PBMC with the optimized IFN-γ ELISpot assay represents a highly standardized, valuable tool to monitor the functionality of CMV-specific CMI with great sensitivity and reliability. Electronic supplementary material The online version of this article (doi:10.1186/s12865-017-0195-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sascha Barabas
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany
| | - Theresa Spindler
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany
| | - Richard Kiener
- Institute of Medical Microbiology and Hygiene, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Charlotte Tonar
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany
| | - Tamara Lugner
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany
| | - Julia Batzilla
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany
| | - Hanna Bendfeldt
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany
| | - Anne Rascle
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany
| | - Benedikt Asbach
- Institute of Medical Microbiology and Hygiene, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Ludwig Deml
- Lophius Biosciences GmbH, Am BioPark 13, 93053, Regensburg, Germany. .,Institute of Medical Microbiology and Hygiene, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
6
|
Massilamany C, Krishnan B, Reddy J. Major Histocompatibility Complex Class II Dextramers: New Tools for the Detection of antigen-Specific, CD4 T Cells in Basic and Clinical Research. Scand J Immunol 2015; 82:399-408. [PMID: 26207337 PMCID: PMC4610346 DOI: 10.1111/sji.12344] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/15/2015] [Indexed: 12/19/2022]
Abstract
The advent of major histocompatibility complex (MHC) tetramer technology has been a major contribution to T cell immunology, because tetramer reagents permit detection of antigen-specific T cells at the single-cell level in heterogeneous populations by flow cytometry. However, unlike MHC class I tetramers, the utility of MHC class II tetramers has been less frequently reported. MHC class II tetramers can be used successfully to enumerate the frequencies of antigen-specific CD4 T cells in cells activated in vitro, but their use for ex vivo analyses continues to be a problem, due in part to their activation dependency for binding with T cells. To circumvent this problem, we recently reported the creation of a new generation of reagents called MHC class II dextramers, which were found to be superior to their counterparts. In this review, we discuss the utility of class II dextramers vis-a-vis tetramers, with respect to their specificity and sensitivity, including potential applications and limitations.
Collapse
Affiliation(s)
- C Massilamany
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - B Krishnan
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - J Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
7
|
Vu T, Rahimian A, Stybayeva G, Gao Y, Kwa T, Van de Water J, Revzin A. Reconfigurable microfluidic device with integrated antibody arrays for capture, multiplexed stimulation, and cytokine profiling of human monocytes. BIOMICROFLUIDICS 2015; 9:044115. [PMID: 26339315 PMCID: PMC4529433 DOI: 10.1063/1.4928128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/27/2015] [Indexed: 06/05/2023]
Abstract
Monocytes represent a class of immune cells that play a key role in the innate and adaptive immune response against infections. One mechanism employed by monocytes for sensing foreign antigens is via toll-like receptors (TLRs)-transmembrane proteins that distinguish classes of foreign pathogens, for example, bacteria (TLR4, 5, and 9) vs. fungi (TLR2) vs. viruses (TLR3, 7, and 8). Binding of antigens activates a signaling cascade through TLR receptors that culminate in secretion of inflammatory cytokines. Detection of these cytokines can provide valuable clinical data for drug developers and disease investigations, but this usually requires a large sample volume and can be technically inefficient with traditional techniques such as flow cytometry, enzyme-linked immunosorbent assay, or luminex. This paper describes an approach whereby antibody arrays for capturing cells and secreted cytokines are encapsulated within a microfluidic device that can be reconfigured to operate in serial or parallel mode. In serial mode, the device represents one long channel that may be perfused with a small volume of minimally processed blood. Once monocytes are captured onto antibody spots imprinted into the floor of the device, the straight channel is reconfigured to form nine individually perfusable chambers. To prove this concept, the microfluidic platform was used to capture monocytes from minimally processed human blood in serial mode and then to stimulate monocytes with different TLR agonists in parallel mode. Three cytokines, tumor necrosis factor-α, interleukin (IL)-6, and IL-10, were detected using anti-cytokine antibody arrays integrated into each of the six chambers. We foresee further use of this device in applications such as pediatric immunology or drug/vaccine testing where it is important to balance small sample volume with the need for high information content.
Collapse
Affiliation(s)
- Tam Vu
- Department of Biomedical Engineering, University of California , 451 Health Sciences Drive, Suite 2619, Davis, California 95616, USA
| | - Ali Rahimian
- Department of Biomedical Engineering, University of California , 451 Health Sciences Drive, Suite 2619, Davis, California 95616, USA
| | - Gulnaz Stybayeva
- Department of Biomedical Engineering, University of California , 451 Health Sciences Drive, Suite 2619, Davis, California 95616, USA
| | - Yandong Gao
- Department of Biomedical Engineering, University of California , 451 Health Sciences Drive, Suite 2619, Davis, California 95616, USA
| | - Timothy Kwa
- Department of Biomedical Engineering, University of California , 451 Health Sciences Drive, Suite 2619, Davis, California 95616, USA
| | - Judy Van de Water
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine , 451 Health Sciences Drive, Suite 6510, Davis, California 95616, USA
| | - Alexander Revzin
- Department of Biomedical Engineering, University of California , 451 Health Sciences Drive, Suite 2619, Davis, California 95616, USA
| |
Collapse
|
8
|
Rekers NV, von Herrath MG, Wesley JD. Immunotherapies and immune biomarkers in Type 1 diabetes: A partnership for success. Clin Immunol 2015; 161:37-43. [PMID: 26122172 DOI: 10.1016/j.clim.2015.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/16/2022]
Abstract
The standard of care (SoC) for Type 1 diabetes (T1D) today is much the same as it was in the early 1920s, simply with more insulin options-fast-acting, slow-acting, injectable, and inhalable insulins. However, these well-tolerated treatments only manage the symptoms and complications, but do nothing to halt the underlying immune response. There is an unmet need for better treatment options for T1D that address all aspects of the disease. For decades, we have successfully treated T1D in preclinical animal models with immune-modifying therapies that have not demonstrated comparable efficacy in humans. The path to bringing such options to the clinic will depend on the implementation and standard inclusion of biomarkers of immune and therapeutic efficacy in T1D clinical trials, and dictate if we can create a new SoC that treats the underlying autoimmunity as well as the symptoms it causes.
Collapse
Affiliation(s)
- Niels V Rekers
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA; Pacific Northwest Diabetes Research Institute, Seattle, WA, USA
| | | | - Johnna D Wesley
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA.
| |
Collapse
|
9
|
Tischer S, Dieks D, Sukdolak C, Bunse C, Figueiredo C, Immenschuh S, Borchers S, Stripecke R, Maecker-Kolhoff B, Blasczyk R, Eiz-Vesper B. Evaluation of suitable target antigens and immunoassays for high-accuracy immune monitoring of cytomegalovirus and Epstein–Barr virus-specific T cells as targets of interest in immunotherapeutic approaches. J Immunol Methods 2014; 408:101-13. [DOI: 10.1016/j.jim.2014.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/12/2014] [Accepted: 05/20/2014] [Indexed: 12/18/2022]
|
10
|
Côté-Bigras S, Dionne A, Asselin-Mullen P, Leblicq C, Rottembourg D. Interferon-gamma ELISPOT detecting reactivity of T cells to TSH receptor peptides in Graves' disease. Clin Endocrinol (Oxf) 2014; 80:296-300. [PMID: 23734883 DOI: 10.1111/cen.12257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 04/24/2013] [Accepted: 05/30/2013] [Indexed: 11/27/2022]
Abstract
OBJECTIVE While thyrotrophin receptor (TSHR) is recognized as the main autoantigen in Graves' disease (GD), the actual antigen specificity of T cells that infiltrate the thyroid and the orbit is unknown. Identifying T cell responses to TSHR peptides has been difficult in the past due to the low frequency of autoreactive T cells and to the diversity of the putative epitopes identified by proliferation assays. METHODS We used the interferon-gamma ELISPOT assay to identify T cell reactivity to TSHR peptides in patients with GD. Peripheral blood T cells were exposed in vitro to four pools of 10 overlapping TSHR peptides. RESULTS T cells from 11 of 31 (35%) patients with GD and 1 of 22 (4%) healthy controls reacted to at least one peptide pool (P = 0·009). Mean time since diagnosis was 3·2 years in responder patients and 5·6 years in nonresponders (P = 0·07). In two patients, T cell reactivity was observed shortly after radioiodine treatment and not thereafter. CONCLUSIONS Our findings demonstrate that the ELISPOT assay is effective to test T cell reactivity in patients with GD and that patients with GD have significantly more interferon-gamma responses towards TSHR peptides than controls. The data suggest that screening for T cell responses in patients with GD might be more efficient in recent-onset disease or after radioiodine treatment.
Collapse
Affiliation(s)
- Sarah Côté-Bigras
- Faculty of Medicine, University Hospital Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | | | |
Collapse
|
11
|
Diagnostic criteria of latent autoimmune diabetes in adults (LADA): a review and reflection. Front Med 2012; 6:243-7. [PMID: 22843304 DOI: 10.1007/s11684-012-0201-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 04/01/2012] [Indexed: 12/11/2022]
Abstract
Diabetes has become a major public health problem in China nowadays. There are almost 97 million diabetic patients nationwide. Latent autoimmune diabetes in adults (LADA) is a subtype of autoimmune diabetes. Although it has been reported for about 20 years, the diagnostic criteria of this disease remain controversial. The discussion mainly focused on serum autoantibodies, period of insulin need and age of diagnosis. Besides, β cell function, metabolic parameters, genetic factors and cell immunity may also contribute to the formulation of the criteria. Here, we aim to review and discuss the diagnostic criteria of latent autoimmune diabetes in adults.
Collapse
|
12
|
Jin A, Ozawa T, Tajiri K, Obata T, Kishi H, Muraguchi A. Rapid isolation of antigen-specific antibody-secreting cells using a chip-based immunospot array. Nat Protoc 2011; 6:668-76. [PMID: 21527923 DOI: 10.1038/nprot.2011.322] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Here we report a new method for isolating antigen-specific antibody-secreting cells (ASCs) using a microwell array chip, which offers a rapid, efficient and high-throughput (up to 234,000 individual cells) system for the detection and retrieval of cells that secrete antibodies of interest on a single-cell basis. We arrayed a large population of lymphoid cells containing ASCs from human peripheral blood on microwell array chips and detected spots with secreted antibodies. This protocol can be completed in less than 7 h, including 3 h of cell culture. The method presented here not only has high sensitivity and specificity comparable with enzyme-linked immunospot (ELISPOT) but it also overcomes the limitations of ELISPOT in recovering ASCs that can be used to produce antigen-specific human monoclonal antibodies. This method can also be used to detect cells secreting molecules other than antibodies, such as cytokines, and it provides a tool for cell analysis and clinical diagnosis.
Collapse
Affiliation(s)
- Aishun Jin
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | | | | | | | | | | |
Collapse
|
13
|
Dang M, Rockell J, Wagner R, Wenzlau JM, Yu L, Hutton JC, Gottlieb PA, Davidson HW. Human type 1 diabetes is associated with T cell autoimmunity to zinc transporter 8. THE JOURNAL OF IMMUNOLOGY 2011; 186:6056-63. [PMID: 21471440 DOI: 10.4049/jimmunol.1003815] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recently we demonstrated that zinc transporter 8 (ZnT8) is a major target of autoantibodies in human type 1 diabetes (T1D). Because the molecules recognized by T1D autoantibodies are typically also targets of autoreactive T cells, we reasoned that this would likely be the case for ZnT8. To test this hypothesis, IFN-γ-producing T cells specific for ZnT8 in the peripheral blood of 35 patients with T1D (<6 mo after onset at blood draw) and 41 age-matched controls were assayed by ELISPOT using a library of 23 overlapping dipeptide pools covering the entire 369 aa primary sequence. Consistent with our hypothesis, patients showed significantly higher T cell reactivity than the matched controls, manifest in terms of the breadth of the overall response and the magnitude of responses to individual pools. Therefore, the median number of pools giving positive responses (stimulation index ≥ 3) in the control group was 1.0 (range, 0-7) compared with 6.0 (range, 1-20; p < 0.0001) for the patients. Similarly, the median stimulation index of positive responses in controls was 3.1 versus 5.0 in the patients (p < 0.0001). Individually, 7 of 23 pools showed significant disease association (p < 0.001), with several of the component peptides binding the disease associated HLA-DR3 (0301) and -DR4 (0401) molecules in vitro. We conclude that ZnT8 is also a major target of disease-associated autoreactive T cells in human T1D, and we suggest that reagents that target ZnT8-specific T cells could have therapeutic potential in preventing or arresting the progression of this disease.
Collapse
Affiliation(s)
- MyLinh Dang
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Mannering SI, Wong FS, Durinovic-Belló I, Brooks-Worrell B, Tree TI, Cilio CM, Schloot NC, Mallone R. Current approaches to measuring human islet-antigen specific T cell function in type 1 diabetes. Clin Exp Immunol 2010; 162:197-209. [PMID: 20846160 DOI: 10.1111/j.1365-2249.2010.04237.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by the T cell-mediated destruction of the pancreatic insulin-producing beta cells. Currently there are no widely accepted and standardized assays available to analyse the function of autoreactive T cells involved in T1D. The development of such an assay would greatly aid efforts to understand the pathogenesis of T1D and is also urgently required to guide the development of antigen-based therapies intended to prevent, or cure, T1D. Here we describe some of the assays used currently to detect autoreactive T cells in human blood and review critically their strengths and weaknesses. The challenges and future prospects for the T cell assays are discussed.
Collapse
Affiliation(s)
- S I Mannering
- St Vincent's Institute, The University of Melbourne, Department of Medicine, St Vincent's Hospital, Fitzroy, Vic, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Afonso G, Scotto M, Renand A, Arvastsson J, Vassilieff D, Cilio CM, Mallone R. Critical parameters in blood processing for T-cell assays: Validation on ELISpot and tetramer platforms. J Immunol Methods 2010; 359:28-36. [PMID: 20641145 DOI: 10.1016/j.jim.2010.05.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
Mannering SI, Brodnicki TC. Recent insights into CD4+ T-cell specificity and function in type 1 diabetes. Expert Rev Clin Immunol 2010; 3:557-64. [PMID: 20477160 DOI: 10.1586/1744666x.3.4.557] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Type 1 diabetes (T1D) is caused by T-cell-mediated destruction of the insulin-producing beta-cells in the pancreas. Genetic and immunological evidence from humans and mouse models indicates that CD4(+) T cells play a crucial role in the development and prevention of T1D. The dichotomy between CD4(+) T regulatory and effector T cells has encouraged research into the role of these cell subsets in T1D. New antigens and epitopes recognized by CD4(+) T cells in affected individuals have been identified. Growing knowledge of T-cell specificity and function is helping to develop new assays for analyzing islet antigen-specific CD4(+) T cells from human blood. Here we discuss, with particular reference to human studies, advances in our understanding of CD4(+) T-cell responses in T1D.
Collapse
Affiliation(s)
- Stuart I Mannering
- Autoimmunity & Transplantation Division, The Walter & Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia.
| | | |
Collapse
|
17
|
Pfleger C, Meierhoff G, Kolb H, Schloot NC. Association of T-cell reactivity with β-cell function in recent onset type 1 diabetes patients. J Autoimmun 2010; 34:127-35. [DOI: 10.1016/j.jaut.2009.08.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 08/12/2009] [Accepted: 08/13/2009] [Indexed: 01/01/2023]
|
18
|
Th1 responses to beta-amyloid in young humans convert to regulatory IL-10 responses in Down syndrome and Alzheimer's disease. Neurobiol Aging 2008; 31:1732-42. [PMID: 19058879 DOI: 10.1016/j.neurobiolaging.2008.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 08/22/2008] [Accepted: 09/16/2008] [Indexed: 11/21/2022]
Abstract
A beta(1-42)-specific antibodies and T-cell proliferation point to the existence of a memory response to A beta(1-42) in humans. Using ELISPOT, we studied A beta(1-42)-specific T cells in individuals of various ages, and in subjects with Trisomy 21 or Alzheimer's disease. We show for the first time that A beta(1-42)-specific Th1-type T-cell memory is present in young humans, producing high levels of IFN-gamma and IL-2. With increasing age, the production of IFN-gamma and IL-2 decreases but is not discontinued in healthy subjects and is accompanied by a sharp rise in CD4(+) T-cell-derived regulatory IL-10 production. In contrast, individuals with Trisomy 21 and with Alzheimer's disease produce IL-10 only in the absence of any effector cytokine. This signifies a switch from a Th1 effector to an IL-10 mediated regulatory response.
Collapse
|
19
|
Martinuzzi E, Scotto M, Énée E, Brezar V, Ribeil JA, van Endert P, Mallone R. Serum-free culture medium and IL-7 costimulation increase the sensitivity of ELISpot detection. J Immunol Methods 2008; 333:61-70. [DOI: 10.1016/j.jim.2008.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 10/30/2007] [Accepted: 01/08/2008] [Indexed: 10/22/2022]
|
20
|
Vrabelova Z, Kolouskova S, Böhmova K, Faresjö MK, Sumnik Z, Pechova M, Kverka M, Chudoba D, Zacharovova K, Stadlerova G, Pithova P, Hladikova M, Stechova K. Protein microarray analysis as a tool for monitoring cellular autoreactivity in type 1 diabetes patients and their relatives. Pediatr Diabetes 2007; 8:252-60. [PMID: 17850467 DOI: 10.1111/j.1399-5448.2007.00308.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Autoreactive T cells have a crucial role in type 1 diabetes (T1D) pathogenesis. OBJECTIVES The aim of our study was to monitor the in vitro production of cytokines by peripheral blood mononuclear cells (PBMCs) after stimulation with diabetogenic autoantigens. SUBJECTS Ten T1D patients (tested at the time of diagnosis and 6 and 12 months later), 10 first-degree relatives of the T1D patients, and 10 controls underwent the study. METHODS PBMCs were stimulated with glutamic acid decarboxylase 65 (GAD65) amino acids (a.a.) 247-279, 509-528, and 524-543; proinsulin a.a. 9-23; and tyrosine phosphatase (islet antigen-2)/R2 a.a. 853-872. Interleukin (IL)-2, IL-4, IL-5, IL-6, IL-10, IL-13, interferon (IFN)-gamma, tumor necrosis factor beta, transforming growth factor beta1, and granulocyte colony-stimulating factor (GCSF) were analyzed by protein microarray. RESULTS Differences in cytokine(s) poststimulatory and mainly in basal production were observed in all groups. The most prominent findings were in controls, the higher basal levels of IL-2, IL-4, IL-5, IL-13, and GCSF were observed when compared with relatives (p < 0.05, for all). After stimulation in controls, there was a significant decrease in IL-2, IL-13, GCSF, and IFN-gamma (p < 0.05, for all). The group of relatives was the most variable in poststimulatory production. A strong correlation between cytokines production was found but groups differed in this aspect. CONCLUSION By multiplex analysis, it may be possible, for example, to define the risk immunological response pattern among relatives or to monitor the immune response in patients on immune modulation therapy.
Collapse
Affiliation(s)
- Zuzana Vrabelova
- Department of Paediatrics, 2nd Medical Faculty of Charles University, Prague, The Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Weiss L, Zeira M, Reich S, Slavin S, Raz I, Mechoulam R, Gallily R. Cannabidiol arrests onset of autoimmune diabetes in NOD mice. Neuropharmacology 2007; 54:244-9. [PMID: 17714746 PMCID: PMC2270485 DOI: 10.1016/j.neuropharm.2007.06.029] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2007] [Revised: 05/31/2007] [Accepted: 06/21/2007] [Indexed: 12/31/2022]
Abstract
We have previously reported that cannabidiol (CBD) lowers the incidence of diabetes in young non-obese diabetes-prone (NOD) female mice. In the present study we show that administration of CBD to 11-14 week old female NOD mice, which are either in a latent diabetes stage or with initial symptoms of diabetes, ameliorates the manifestations of the disease. Diabetes was diagnosed in only 32% of the mice in the CBD-treated group, compared to 86% and 100% in the emulsifier-treated and untreated groups, respectively. In addition, the level of the proinflammatory cytokine IL-12 produced by splenocytes was significantly reduced, whereas the level of the anti-inflammatory IL-10 was significantly elevated following CBD-treatment. Histological examination of the pancreata of CBD-treated mice revealed more intact islets than in the controls. Our data strengthen our previous assumption that CBD, known to be safe in man, can possibly be used as a therapeutic agent for treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Lola Weiss
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Hebrew University Hospital, Jerusalem 91120, Israel.
| | | | | | | | | | | | | |
Collapse
|
22
|
Lee S, Miller SA, Wright DW, Rock MT, Crowe JE. Tissue-specific regulation of CD8+ T-lymphocyte immunodominance in respiratory syncytial virus infection. J Virol 2006; 81:2349-58. [PMID: 17182672 PMCID: PMC1865932 DOI: 10.1128/jvi.01910-06] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) are critical for control of respiratory syncytial virus (RSV) infection in humans and mice. To investigate cellular immune responses to infection, it is important to identify major histocompatibility complex (MHC) class I-restricted CTL epitopes. In this study, we identified a new RSV-specific, H-2K(d)-restricted subdominant epitope in the M2 protein, M2(127-135) (amino acids 127 to 135). This finding allowed us to study the frequency of T lymphocytes responding to two H-2K(d)-presented epitopes in the same protein following RSV infection by enzyme-linked immunospot (ELISPOT) and intracellular cytokine assays for both lymphoid and nonlymphoid tissues. For the subdominant epitope, we identified an optimal nine-amino-acid peptide, VYNTVISYI, which contained an H-2K(d) consensus sequence with Y at position 2 and I at position 9. In addition, an MHC class I stabilization assay using TAP-2-deficient RMA-S cells transfected with K(d) or L(d) indicated that the epitope was presented by K(d). The ratios of T lymphocytes during the peak CTL response to RSV infection that were specific for M2(82-90) (dominant) to T lymphocytes specific for M2(127-135) (subdominant) were approximately 3:1 in the spleen and 10:1 in the lung. These ratios were observed consistently in primary or secondary infection by the ELISPOT assay and in secondary infection by MHC/peptide tetramer staining. The number of antigen-specific T lymphocytes dropped in the 6 weeks after infection; however, the proportions of T lymphocytes specific for the immunodominant and subdominant epitopes were maintained to a remarkable degree in a tissue-specific manner. These studies will facilitate investigation of the regulation of immunodominance of RSV-specific CTL epitopes.
Collapse
Affiliation(s)
- Sujin Lee
- Department of Pediatrics, Vanderbilt University Medical Center, T-2220 Medical Center North, 1161 21st Avenue South, Nashville, TN 37232-2905, USA
| | | | | | | | | |
Collapse
|
23
|
Zhu P, Li XY, Wang HK, Jia JF, Zheng ZH, Ding J, Fan CM. Oral administration of type-II collagen peptide 250-270 suppresses specific cellular and humoral immune response in collagen-induced arthritis. Clin Immunol 2006; 122:75-84. [PMID: 17045846 DOI: 10.1016/j.clim.2006.08.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Revised: 08/04/2006] [Accepted: 08/04/2006] [Indexed: 11/18/2022]
Abstract
Oral antigen is an attractive approach for the treatment of autoimmune and inflammatory diseases. Establishment of immune markers and methods in evaluating the effects of antigen-specific cellular and humoral immune responses will help the application of oral tolerance in the treatment of human diseases. The present article observed the effects of chicken collagen II (CII), the recombinant polymerized human collagen II 250-270 (rhCII 250-270) peptide and synthesized human CII 250-270 (syCII 250-270) peptide on the induction of antigen-specific autoimmune response in rheumatoid arthritis (RA) peripheral blood mononuclear cells (PBMC) and on the specific cellular and humoral immune response in collagen-induced arthritis (CIA) and mice fed with CII (250-270) prior to immunization with CII. In the study, proliferation, activation and intracellular cytokine production of antigen-specific T lymphocytes were simultaneously analyzed by bromodeoxyuridine (BrdU) incorporation and flow cytometry at the single-cell level. The antigen-specific antibody and antibody-forming cells were detected by ELISA and ELISPOT, respectively. CII (250-270) was found to have stimulated the response of specific lymphocytes in PBMC from RA patients, including the increase expression of surface activation antigen marker CD69 and CD25, and DNA synthesis. Mice, fed with CII (250-270) before CII immunization, had significantly lower arthritic scores than the mice immunized with CII alone, and the body weight of the former increased during the study period. Furthermore, the specific T cell activity, proliferation and secretion of interferon (IFN)-gamma in spleen cells were actively suppressed in CII (250-270)-fed mice, and the serum anti-CII, anti-CII (250-270) antibody activities and the frequency of specific antibody-forming spleen cells were significantly lower in CII (250-270)-fed mice than in mice immunized with CII alone. These observations suggest that oral administration of CII (250-270) can suppress the cellular and humoral immune response in collagen-induced arthritis, and the simultaneous analysis of antigen-specific cellular and humoral immune responses at single-cell level will help the understanding of the oral tolerance mechanisms in CIA and the development of innovative therapeutic intervention for RA.
Collapse
Affiliation(s)
- Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, PR China.
| | | | | | | | | | | | | |
Collapse
|
24
|
Alleva DG, Maki RA, Putnam AL, Robinson JM, Kipnes MS, Dandona P, Marks JB, Simmons DL, Greenbaum CJ, Jimenez RG, Conlon PJ, Gottlieb PA. Immunomodulation in type 1 diabetes by NBI-6024, an altered peptide ligand of the insulin B epitope. Scand J Immunol 2006; 63:59-69. [PMID: 16398702 DOI: 10.1111/j.1365-3083.2005.01705.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
NBI-6024 is an altered peptide ligand (APL) corresponding to the 9-23 amino acid region of the insulin B chain (B(9-23)), an epitope recognized by inflammatory interferon-gamma-producing T helper (Th)1 lymphocytes in type 1 diabetic patients. Immunomodulatory effects of NBI-6024 administration in recent-onset diabetic patients in a phase I clinical trial (NBI-6024-0003) were measured in peripheral blood mononuclear cells using the enzyme-linked immunosorbent spot assay. Analysis of the mean magnitude of cytokine responses to B(9-23) and NBI-6024 for each cohort showed significant increases in interleukin-5 responses (a Th2 regulatory phenotype) in cohorts that received APL relative to those receiving placebo. A responder analysis showed that Th1 responses to B(9-23) and NBI-6024 were observed almost exclusively in the placebo-treated diabetic population but not in nondiabetic control subjects and that APL administration (five biweekly subcutaneous injections) significantly and dose-dependently reduced the percentage of patients with these Th1 responses. The results of this phase I clinical study strongly suggest that NBI-6024 treatment shifted the Th1 pathogenic responses in recent-onset type 1 diabetic patients to a protective Th2 regulatory phenotype. The significance of these findings on the clinical outcome of disease is currently under investigation in a phase II multidose study.
Collapse
Affiliation(s)
- D G Alleva
- Neurocrine Biosciences, Inc., San Diego, CA 92130, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Tassignon J, Burny W, Dahmani S, Zhou L, Stordeur P, Byl B, De Groote D. Monitoring of cellular responses after vaccination against tetanus toxoid: comparison of the measurement of IFN-gamma production by ELISA, ELISPOT, flow cytometry and real-time PCR. J Immunol Methods 2005; 305:188-98. [PMID: 16157348 DOI: 10.1016/j.jim.2005.07.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 05/31/2005] [Accepted: 07/19/2005] [Indexed: 11/23/2022]
Abstract
One of the challenges for immunomonitoring in clinical trials is to detect an antigen specific T cell-mediated immune response. In an attempt to define the most suitable assay, tetanus toxoid was used to compare the capacity of 4 different methods to detect cytokine responses, before and after recall vaccination, in peripheral blood mononuclear cells (PBMC) of 14 healthy volunteers. ELISA, ELISPOT, intracytoplasmic detection and real-time RT-PCR were chosen to measure IFN-gamma production before and after vaccination. As far as the detection of memory T cell status (before vaccination) was concerned, we found that ELISPOT was the most sensitive method to discriminate TT-induced from spontaneous responses. On the other hand, intracytoplasmic cytokine detection was the most efficient method to detect the restimulating effect of TT vaccination.
Collapse
|
26
|
Turner CK, Blieden TM, Smith TJ, Feldon SE, Foster DC, Sime PJ, Phipps RP. A novel ELISpot method for adherent cells. J Immunol Methods 2004; 291:63-70. [PMID: 15345305 DOI: 10.1016/j.jim.2004.04.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2004] [Revised: 04/28/2004] [Accepted: 04/28/2004] [Indexed: 11/17/2022]
Abstract
The purpose of this study was to develop an enzyme-linked immunospot assay (ELISpot assay) that can be used with human adherent cells. While standard enzyme-linked immunosorbent assays (ELISAs) are available and widely used and ELISpot assays are used for nonadherent lymphocytes, no ELISpot assay has been developed for adherent cells. We used primary human fibroblasts from four different tissues (myometrium, lung, gingiva, and orbit), either unstimulated or interleukin (IL)-1beta-activated, to evaluate an ELISpot assay. Antibody pairs for IL-6 and IL-8 were used and results were compared to a standard ELISA. We found that we could reliably detect IL-6 and IL-8 spots with as few as 10 fibroblasts. Optimal cell numbers were 50 cells per well incubated for 8 h, although spots appeared as early as 2 h after incubation. Spots were absent when cells, primary, or secondary anti-cytokine antibodies were omitted from the protocol. Spot number and size can be ascertained using current automated ELISpot reader technology. The frequency of IL-6 and IL-8-producing human fibroblasts could also be determined. For example, 60% of the lung fibroblasts express IL-6, but IL-8 can be detected from only 40% of the cells. Approximately 80% of the human orbital fibroblasts make IL-6, whereas approximately 50% generate IL-8 following IL-1beta stimulation. These new findings show that fibroblasts from different human tissues display different frequencies of cytokine production and this further supports the concept of fibroblast diversity. The sensitivity of this new ELISpot assay is adequate for cytokine detection in just a few cells, unlike the standard ELISA. It should permit ascertaining the frequency of fibroblasts and other adherent cells that produce cytokines and, if desired, can be used in tandem with a standard ELISA to determine total cytokine produced. Moreover, the assay is suitable for normal human adherent cells that are often short-lived and difficult to cultivate.
Collapse
Affiliation(s)
- Chantal K Turner
- Department of Environmental Medicine, University of Rochester, 601 Elmwood Ave, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Arif S, Tree TI, Astill TP, Tremble JM, Bishop AJ, Dayan CM, Roep BO, Peakman M. Autoreactive T cell responses show proinflammatory polarization in diabetes but a regulatory phenotype in health. J Clin Invest 2004; 113:451-63. [PMID: 14755342 PMCID: PMC324541 DOI: 10.1172/jci19585] [Citation(s) in RCA: 359] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2003] [Accepted: 11/25/2003] [Indexed: 01/28/2023] Open
Abstract
According to the quality of response they mediate, autoreactive T cells recognizing islet beta cell peptides could represent both disease effectors in the development of type 1 diabetes (T1DM) and directors of tolerance in nondiabetic individuals or those undergoing preventative immunotherapy. A combination of the rarity of these cells, inadequate technology, and poorly defined epitopes, however, has hampered examination of this paradigm. We have identified a panel of naturally processed islet epitopes by direct elution from APCs bearing HLA-DR4. Employing these epitopes in a sensitive, novel cytokine enzyme-linked immunosorbent spot assay, we show that the quality of autoreactive T cells in patients with T1DM exhibits extreme polarization toward a proinflammatory Th1 phenotype. Furthermore, we demonstrate that rather than being unresponsive, the majority of nondiabetic, HLA-matched control subjects also manifest a response against islet peptides, but one that shows extreme T regulatory cell (Treg, IL-10-secreting) bias. We conclude that development of T1DM depends on the balance of autoreactive Th1 and Treg cells, which may be open to favorable manipulation by immune intervention.
Collapse
Affiliation(s)
- Sefina Arif
- Department of Immunobiology, Guy's, King's and St. Thomas' School of Medicine, King's College London, Guy's Campus, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Schloot NC, Meierhoff G, Karlsson Faresjö M, Ott P, Putnam A, Lehmann P, Gottlieb P, Roep BO, Peakman M, Tree T. Comparison of cytokine ELISpot assay formats for the detection of islet antigen autoreactive T cells. J Autoimmun 2003; 21:365-76. [PMID: 14624759 DOI: 10.1016/s0896-8411(03)00111-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The identification of sensitive assay formats capable of distinguishing islet autoreactive T cells directly ex vivo in blood is a major goal in type 1 diabetes research. Recently, much interest has been shown in the cytokine enzyme linked immunospot assay (CK ELISpot), an assay potentially capable of fulfilling these difficult criteria. To address the utility of this assay in detecting autoreactive T cells, a 'wet' workshop was organized using the same fresh blood sample and coded antigens. Five different laboratories participated, using three distinct CK ELISpot assay formats. Samples from two subjects were pre-tested for responses to sub-optimal concentrations of tetanus toxoid, representing a low frequency recall response, and peptides from diabetes associated autoantigens GAD65, IA-2 and HSP60. All participants measured interferon-gamma production and combinations of interleukins-4, -5, -10 and -13. In the workshop 4 of 5 laboratories detected low frequency recall responses in both subjects and 3 of 5 detected at least one of the autoreactive peptide responses concordant with pre-testing. Significant assay format related differences in sensitivity and signal-to-noise ratio were observed. The results demonstrate the potential for detection of low-level autoreactive T cell responses and identify assay characteristics that will be useful for studies in type 1 diabetes.
Collapse
Affiliation(s)
- Nanette C Schloot
- German Diabetes Research Institute at the Heinrich Heine University, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Current literature in diabetes. Diabetes Metab Res Rev 2003; 19:164-71. [PMID: 12673786 DOI: 10.1002/dmrr.347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|