1
|
Golikova EA, Alshevskaya AA, Alrhmoun S, Sivitskaya NA, Sennikov SV. TCR-T cell therapy: current development approaches, preclinical evaluation, and perspectives on regulatory challenges. J Transl Med 2024; 22:897. [PMID: 39367419 PMCID: PMC11451006 DOI: 10.1186/s12967-024-05703-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
TCR-T cell therapy represents a promising advancement in adoptive immunotherapy for cancer treatment. Despite its potential, the development and preclinical testing of TCR-T cells face significant challenges. This review provides a structured overview of the key stages in preclinical testing, including in silico, in vitro, and in vivo methods, within the context of the sequential development of novel therapies. This review aimed to systematically outline the processes for evaluating TCR-T cells at each stage: from in silico approaches used to predict target antigens, assess cross-reactivity, and minimize off-target effects, to in vitro assays designed to measure cell functionality, cytotoxicity, and activation. Additionally, the review discusses the limitations of in vivo testing in animal models, particularly in accurately reflecting the human tumor microenvironment and immune responses. Performed analysis emphasizes the importance of these preclinical stages in the safe and effective development of TCR-T cell therapies. While current models provide valuable insights, we identify critical gaps, particularly in in vivo biodistribution and toxicity assessments, and propose the need for enhanced standardization and the development of more representative models. This structured approach aims to improve the predictability and safety of TCR-T cell therapy as it advances towards clinical application.
Collapse
Affiliation(s)
- Elena A Golikova
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
| | - Alina A Alshevskaya
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia.
| | - Saleh Alrhmoun
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
- Federal State Budgetary Scientific Institution, "Research Institute of Fundamental and Clinical Immunology" (RIFCI), 630099, Novosibirsk, Russia
| | - Natalia A Sivitskaya
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
| | - Sergey V Sennikov
- Federal State Autonomous Educational Institution of Higher Education, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119435, Moscow, Russia
- Federal State Budgetary Scientific Institution, "Research Institute of Fundamental and Clinical Immunology" (RIFCI), 630099, Novosibirsk, Russia
| |
Collapse
|
2
|
Tiwari R, Singh VK, Rajneesh, Kumar A, Gautam V, Kumar R. MHC tetramer technology: Exploring T cell biology in health and disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:327-345. [PMID: 38762273 DOI: 10.1016/bs.apcsb.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Major histocompatibility complex (MHC) tetramers stand as formidable tools within T cell biology, facilitating the exploration and comprehension of immune responses. These artificial molecules, comprising four bound MHC molecules, typically with a specified peptide and a fluorescent label, play a pivotal role in characterizing T cell subsets, monitoring clonal expansion, and unraveling T cell dynamics during responses to infections or immunotherapies. Beyond their applications in T cell biology, MHC tetramers prove valuable in investigating a spectrum of diseases such as infectious diseases, autoimmune disorders, and cancers. Their instrumental role extends to vaccine research and development. Notably, when appropriately configured, tetramers transcend T cell biology research and find utility in exploring natural killer T cells and contributing to specific T cell clonal deletions.
Collapse
Affiliation(s)
- Rahul Tiwari
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajneesh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
3
|
Vasoya D, Connelley T, Tzelos T, Todd H, Ballingall KT. Large scale transcriptional analysis of MHC class I haplotype diversity in sheep. HLA 2024; 103:e15356. [PMID: 38304958 DOI: 10.1111/tan.15356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/15/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
Domestic sheep (Ovis aries) have been an important component of livestock agricultural production for thousands of years. Preserving genetic diversity within livestock populations maintains a capacity to respond to changing environments and rapidly evolving pathogens. MHC genetic diversity can influence immune functionality at individual and population levels. Here, we focus on defining functional MHC class I haplotype diversity in a large cohort of Scottish Blackface sheep pre-selected for high levels of MHC class II DRB1 diversity. Using high-throughput amplicon sequencing with three independent sets of barcoded primers we identified 134 MHC class I transcripts within 38 haplotypes. Haplotypes were identified with between two and six MHC class I genes, plus variable numbers of conserved sequences with very low read frequencies. One or two highly transcribed transcripts dominate each haplotype indicative of two highly polymorphic, classical MHC class I genes. Additional clusters of medium, low, and very low expressed transcripts are described, indicative of lower transcribed classical, non-classical and genes whose function remains to be determined.
Collapse
Affiliation(s)
- Deepali Vasoya
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Scotland, UK
| | - Timothy Connelley
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Scotland, UK
| | - Thomas Tzelos
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Scotland, UK
- Moredun Research Institute, Pentlands Science Park, Scotland, UK
| | - Helen Todd
- Moredun Research Institute, Pentlands Science Park, Scotland, UK
| | | |
Collapse
|
4
|
Tippalagama R, Chihab LY, Kearns K, Lewis S, Panda S, Willemsen L, Burel JG, Lindestam Arlehamn CS. Antigen-specificity measurements are the key to understanding T cell responses. Front Immunol 2023; 14:1127470. [PMID: 37122719 PMCID: PMC10140422 DOI: 10.3389/fimmu.2023.1127470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Antigen-specific T cells play a central role in the adaptive immune response and come in a wide range of phenotypes. T cell receptors (TCRs) mediate the antigen-specificities found in T cells. Importantly, high-throughput TCR sequencing provides a fingerprint which allows tracking of specific T cells and their clonal expansion in response to particular antigens. As a result, many studies have leveraged TCR sequencing in an attempt to elucidate the role of antigen-specific T cells in various contexts. Here, we discuss the published approaches to studying antigen-specific T cells and their specific TCR repertoire. Further, we discuss how these methods have been applied to study the TCR repertoire in various diseases in order to characterize the antigen-specific T cells involved in the immune control of disease.
Collapse
|
5
|
Ide H, Aoshi T, Saito M, Espulgar WV, Briones JC, Hosokawa M, Matsunaga H, Arikawa K, Takeyama H, Koyama S, Takamatsu H, Tamiya E. Linking antigen specific T-cell dynamics in a microfluidic chip to single cell transcription patterns. Biochem Biophys Res Commun 2023; 657:8-15. [PMID: 36963175 DOI: 10.1016/j.bbrc.2023.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
A new non-invasive screening profile has been realized that can aid in determining T-cell activation state at single-cell level. Production of activated T-cells with good specificity and stable proliferation is greatly beneficial for advancing adoptive immunotherapy as innate immunological cells are not effective in recognizing and eliminating cancer as expected. The screening method is realized by relating intracellular Ca2+ intensity and motility of T-cells interacting with APC (Antigen Presenting Cells) in a microfluidic chip. The system is tested using APC pulsed with OVA257-264 peptide and its modified affinities (N4, Q4, T4 and V4), and the T-cells from OT-1 mice. In addition, single cell RNA sequencing reveals the activation states of the cells and the clusters from the derived profiles can be indicative of the T-cell activation state. The presented system here can be versatile for a comprehensive application to proceed with T-cell-based immunotherapy and screen the antigen-specific T-cells with excellent efficiency and high proliferation.
Collapse
Affiliation(s)
- Hiroki Ide
- Graduate School of Engineering Osaka Univ, Japan; PhotoBIO Lab, AIST-Osaka Univ, Japan
| | - Taiki Aoshi
- Research Institute for Microbial Diseases, Osaka Univ, Japan
| | - Masato Saito
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
| | | | - Jonathan Campos Briones
- Life and Medical Photonics Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Masahito Hosokawa
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Koji Arikawa
- Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda Univ, Japan; CBBD-OIL, AIST-Waseda Univ, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda Univ, Japan; Research Organization for Nano and Life Innovation, Waseda Univ, Japan
| | | | | | - Eiichi Tamiya
- PhotoBIO Lab, AIST-Osaka Univ, Japan; Institute of Scientific and Industrial Research, Osaka University, Japan
| |
Collapse
|
6
|
Yang G, Wang J, Sun P, Qin J, Yang X, Chen D, Zhang Y, Zhong N, Wang Z. SARS-CoV-2 epitope-specific T cells: Immunity response feature, TCR repertoire characteristics and cross-reactivity. Front Immunol 2023; 14:1146196. [PMID: 36969254 PMCID: PMC10036809 DOI: 10.3389/fimmu.2023.1146196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
The devastating COVID-19 pandemic caused by SARS-CoV-2 and multiple variants or subvariants remains an ongoing global challenge. SARS-CoV-2-specific T cell responses play a critical role in early virus clearance, disease severity control, limiting the viral transmission and underpinning COVID-19 vaccine efficacy. Studies estimated broad and robust T cell responses in each individual recognized at least 30 to 40 SARS-CoV-2 antigen epitopes and associated with COVID-19 clinical outcome. Several key immunodominant viral proteome epitopes, including S protein- and non-S protein-derived epitopes, may primarily induce potent and long-lasting antiviral protective effects. In this review, we summarized the immune response features of immunodominant epitope-specific T cells targeting different SRAS-CoV-2 proteome structures after infection and vaccination, including abundance, magnitude, frequency, phenotypic features and response kinetics. Further, we analyzed the epitopes immunodominance hierarchy in combination with multiple epitope-specific T cell attributes and TCR repertoires characteristics, and discussed the significant implications of cross-reactive T cells toward HCoVs, SRAS-CoV-2 and variants of concern, especially Omicron. This review may be essential for mapping the landscape of T cell responses toward SARS-CoV-2 and optimizing the current vaccine strategy.
Collapse
Affiliation(s)
- Gang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Guangzhou Laboratory, Guangzhou, China
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Junxiang Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ping Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Jian Qin
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xiaoyun Yang
- Guangzhou Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Daxiang Chen
- Guangzhou Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yunhui Zhang
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Nanshan Zhong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Guangzhou Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Zhongfang Wang
- Guangzhou Laboratory, Guangzhou, China
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Cotton AD, Wells JA, Seiple IB. Biotin as a Reactive Handle to Selectively Label Proteins and DNA with Small Molecules. ACS Chem Biol 2022; 17:3270-3275. [PMID: 34410115 DOI: 10.1021/acschembio.1c00252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Biotin is a common functional handle for bioconjugation to proteins and DNA, but its uses are limited to protein-containing conjugation partners such as streptavidin and derivatives thereof. Recently, oxaziridine reagents were developed that selectively conjugate the thioether of methionines on the surface of proteins, a method termed redox-activated chemical tagging (ReACT). These reagents generate sulfimide linkages that range in stability depending on the solvent accessibility and substitutions on the oxaziridine. Here we show that oxaziridine reagents react rapidly with the thioether in biotin to produce sulfimide products that are stable for more than 10 d at 37 °C. This method, which we call biotin redox-activated chemical tagging (BioReACT), expands the utility of biotin labeling and enables a predictable and stable chemical conjugation to biomolecules without the need to screen for a suitable methionine conjugation site. We demonstrate the versatility of this approach by producing a fluorescently labeled antibody, an antibody-drug conjugate, and a small molecule-conjugated oligonucleotide. We anticipate that BioReACT will be useful to rapidly introduce biorthogonal handles into biomolecules using biotin, a functional group that is widespread and straightforward to install.
Collapse
Affiliation(s)
- Adam D Cotton
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco 94143, California, United States
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco 94143, California, United States
| | - Ian B Seiple
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco 94143, California, United States.,Cardiovascular Research Institute, University of California San Francisco, San Francisco 94143, California, United States
| |
Collapse
|
8
|
Targeting the CD47-SIRPα Axis: Present Therapies and the Future for Cutaneous T-cell Lymphoma. Cells 2022; 11:cells11223591. [PMID: 36429020 PMCID: PMC9688096 DOI: 10.3390/cells11223591] [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: 09/05/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The loss of CD47 on aging cells serves as a signal to macrophages to eliminate the target. Therefore, CD47 is a "do-not-eat-me" sign preventing macrophagal phagocytosis via interaction with its ligand SIRPα. Malignant lymphocytes of mycosis fungoides and Sézary syndrome express CD47 highly, thus, being ideal candidates for targeted anti-CD47 therapies. The classes of current anti-CD47-SIRPα therapeutic molecules present in a large variety and include monoclonal antibodies against CD47 and SIRPα, bioengineered SIRPα proteins, miRNAs, and bispecific antibodies. We provided a detailed analysis of all available investigational drugs in a contest of cutaneous T-cell lymphoma. A combination of blockade of the CD47-SIRPα axis and secondary targets in the tumor microenvironment (TME) may improve the clinical efficacy of current immunotherapeutic approaches. We evaluated the possible combination and outlined the most promising one.
Collapse
|
9
|
Patel S, Ramnoruth N, Wehr P, Rossjohn J, Reid HH, Campbell K, Nel HJ, Thomas R. Evaluation of a fit-for-purpose assay to monitor antigen-specific functional CD4+ T-cell subpopulations in rheumatoid arthritis using flow cytometry-based peptide-MHC class-II tetramer staining. Clin Exp Immunol 2022; 207:72-83. [PMID: 35020859 PMCID: PMC8802177 DOI: 10.1093/cei/uxab008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/13/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Antigen-specific T cells can serve as a response biomarker in non-clinical or clinical immunotherapy studies in autoimmune disease. There are protocols with optimized multimer staining methods to detect peptide (p)MHCII+ CD4+ T cells, and some qualified and validated protocols for pMHCI+ CD8+ T cells. However, no protocol is fully or partially qualified to enumerate and characterize antigen-specific pMHCII+ CD4+ T cells from patient samples. Implementing such an assay requires a desired level of specificity and precision, in terms of assay repeatability and reproducibility. In transgenic type II collagen (CII)-immunized HLA-DR1/DR4 humanized mouse models of collagen-induced arthritis (CIA), CII259-273-specific T cells dominantly expand. Therefore antigen-specific T cells recognizing this epitope presented by rheumatoid arthritis (RA)-associated risk HLA-DR allomorphs are of interest to understand disease progression and responses to immunotherapy in RA patients. Using HLA-DRB1∗04:01 or ∗01:01-collagen type II (CII)259-273 tetramers, we evaluated parameters influencing precision and reproducibility of an optimized flow cytometry-based method for antigen-specific CD4+ T cells and eight specific subpopulations with and without tetramer positivity. We evaluated specificity, precision, and reproducibility for research environments and non-regulated laboratories. The assay has excellent overall precision with %CV<25% for intra-assay repeatability, inter-analyst precision, and inter-assay reproducibility. The precision of the assay correlated negatively with the cell viability after thawing, indicating that post-thaw viability is a critical parameter for reproducibility. This assay is suitable for longitudinal analysis of treatment response and disease activity outcome in RA patients, and adaptable for translational or immunotherapy clinical trial settings.
Collapse
Affiliation(s)
- Swati Patel
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Nishta Ramnoruth
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Pascale Wehr
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Hugh H Reid
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, VIC, Australia
| | - Kim Campbell
- Janssen Research & Development, LLC, Spring House, PA, USA
- Janssen Research & Development, LLC, La Jolla, CA, USA
| | - Hendrik J Nel
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
10
|
Jivanjee T, Ibrahim S, Nyquist SK, Gatter GJ, Bromley JD, Jaiswal S, Berger B, Behar SM, Love JC, Shalek AK. Enriching and Characterizing T Cell Repertoires from 3' Barcoded Single-Cell Whole Transcriptome Amplification Products. Methods Mol Biol 2022; 2574:159-182. [PMID: 36087201 DOI: 10.1007/978-1-0716-2712-9_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Antigen-specific T cells play an essential role in immunoregulation and many diseases such as cancer. Characterizing the T cell receptor (TCR) sequences that encode T cell specificity is critical for elucidating the antigenic determinants of immunological diseases and designing therapeutic remedies. However, methods of obtaining single-cell TCR sequencing data are labor and cost intensive, typically requiring both cell sorting and full-length single-cell RNA-sequencing (scRNA-seq). New high-throughput 3' cell-barcoding scRNA-seq methods can simplify and scale this process; however, they do not routinely capture TCR sequences during library preparation and sequencing. While 5' cell-barcoding scRNA-seq methods can be used to examine TCR repertoire at single-cell resolution, doing so requires specialized reagents which cannot be applied to samples previously processed using 3' cell-barcoding methods.Here, we outline a method for sequencing TCRα and TCRβ transcripts from samples already processed using 3' cell-barcoding scRNA-seq platforms, ensuring TCR recovery at a single-cell resolution. In short, a fraction of the 3' barcoded whole transcriptome amplification (WTA) product typically used to generate a massively parallel 3' scRNA-seq library is enriched for TCR transcripts using biotinylated probes and further amplified using the same universal primer sequence from WTA. Primer extension using TCR V-region primers and targeted PCR amplification using a second universal primer result in a 3' barcoded single-cell CDR3-enriched library that can be sequenced with custom sequencing primers. Coupled with 3' scRNA-seq of the same WTA, this method enables simultaneous analysis of single-cell transcriptomes and TCR sequences which can help interpret inherent heterogeneity among antigen-specific T cells and salient disease biology. The method presented here can also be adapted readily to enrich and sequence other transcripts of interest from both 3' and 5' barcoded scRNA-seq WTA libraries.
Collapse
Affiliation(s)
- Tasneem Jivanjee
- Institute for Medical Engineering & Science and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samira Ibrahim
- Institute for Medical Engineering & Science and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah K Nyquist
- Institute for Medical Engineering & Science and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - G James Gatter
- Institute for Medical Engineering & Science and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joshua D Bromley
- Institute for Medical Engineering & Science and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA
- Microbiology Graduate Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Swati Jaiswal
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Bonnie Berger
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel M Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Alex K Shalek
- Institute for Medical Engineering & Science and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
11
|
Svitek N, Taracha ELN, Saya R, Awino E, Nene V, Steinaa L. Analysis of the Cellular Immune Responses to Vaccines. Methods Mol Biol 2022; 2465:283-301. [PMID: 35118627 DOI: 10.1007/978-1-0716-2168-4_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Flow cytometry, enzyme-linked immunospot (ELISpot), and cellular cytotoxicity assays are powerful tools for studying the cellular immune response toward intracellular pathogens and vaccines in livestock species. Lymphocytes from immunized animals can be purified using Ficoll-Paque density gradient centrifugation and evaluated for their antigen specificity or reactivity toward a vaccine. Here, we describe staining of bovine lymphocytes with peptide (p)-MHC class I tetramers and antibodies specific toward cellular activation markers for evaluation by multiparametric flow cytometry, as well as interferon (IFN)-γ ELISpot and cytotoxicity using chromium (51Cr) release assays. A small component on the use of immunoinformatics for fine-tuning the identification of a minimal CTL epitope is included, and a newly developed and simple assay to measure TCR avidity.
Collapse
Affiliation(s)
- Nicholas Svitek
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
| | | | - Rosemary Saya
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
| | - Elias Awino
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
| | - Vish Nene
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya
| | - Lucilla Steinaa
- Animal and Human Health, International Livestock Research Institute, Nairobi, Kenya.
| |
Collapse
|
12
|
Vrbensky JR, Nazy I, Clare R, Larché M, Arnold DM. T cell-mediated autoimmunity in immune thrombocytopenia. Eur J Haematol 2021; 108:18-27. [PMID: 34487584 DOI: 10.1111/ejh.13705] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/22/2022]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by a low platelet count and an increased risk of bleeding. In addition to anti-platelet autoantibodies, CD8+ T cells have been implicated as a mechanism of platelet destruction. The current evidence for the existence of platelet-specific CD8+ T cells in ITP is inconclusive. The purpose of this review is to summarize the studies that investigated CD8+ T cells in ITP and to review the methods that have been used to detect autoreactive CD8+ T cells in other autoimmune diseases.
Collapse
Affiliation(s)
- John R Vrbensky
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ishac Nazy
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada.,McMaster Centre for Transfusion Research, McMaster University, Hamilton, ON, Canada
| | - Rumi Clare
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Mark Larché
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Donald M Arnold
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada.,McMaster Centre for Transfusion Research, McMaster University, Hamilton, ON, Canada.,Canadian Blood Services, Hamilton, ON, Canada
| |
Collapse
|
13
|
Vaurs J, Douchin G, Echasserieau K, Oger R, Jouand N, Fortun A, Hesnard L, Croyal M, Pecorari F, Gervois N, Bernardeau K. A novel and efficient approach to high-throughput production of HLA-E/peptide monomer for T-cell epitope screening. Sci Rep 2021; 11:17234. [PMID: 34446788 PMCID: PMC8390762 DOI: 10.1038/s41598-021-96560-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/10/2021] [Indexed: 12/05/2022] Open
Abstract
Over the past two decades, there has been a great interest in the study of HLA-E-restricted αβ T cells during bacterial and viral infections, including recently SARS-CoV-2 infection. Phenotyping of these specific HLA-E-restricted T cells requires new tools such as tetramers for rapid cell staining or sorting, as well as for the identification of new peptides capable to bind to the HLA-E pocket. To this aim, we have developed an optimal photosensitive peptide to generate stable HLA-E/pUV complexes allowing high-throughput production of new HLA-E/peptide complexes by peptide exchange. We characterized the UV exchange by ELISA and improved the peptide exchange readout using size exclusion chromatography. This novel approach for complex quantification is indeed very important to perform tetramerization of MHC/peptide complexes with the high quality required for detection of specific T cells. Our approach allows the rapid screening of peptides capable of binding to the non-classical human HLA-E allele, paving the way for the development of new therapeutic approaches based on the detection of HLA-E-restricted T cells.
Collapse
Affiliation(s)
- Juliette Vaurs
- P2R "Production de Protéines Recombinantes", Université de Nantes, CRCINA, SFR-Santé, INSERM, CNRS, CHU Nantes, Nantes, France
| | - Gaël Douchin
- P2R "Production de Protéines Recombinantes", Université de Nantes, CRCINA, SFR-Santé, INSERM, CNRS, CHU Nantes, Nantes, France
| | - Klara Echasserieau
- P2R "Production de Protéines Recombinantes", Université de Nantes, CRCINA, SFR-Santé, INSERM, CNRS, CHU Nantes, Nantes, France
- Université de Nantes, Inserm, CRCINA, 44000, Nantes, France
| | - Romain Oger
- Université de Nantes, Inserm, CRCINA, 44000, Nantes, France
- LabEx IGO «Immunotherapy, Graft, Oncology», Nantes, France
| | - Nicolas Jouand
- Université de Nantes, Inserm, CRCINA, 44000, Nantes, France
- Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, 44000, Nantes, France
| | - Agnès Fortun
- P2R "Production de Protéines Recombinantes", Université de Nantes, CRCINA, SFR-Santé, INSERM, CNRS, CHU Nantes, Nantes, France
- Université de Nantes, CHU de Nantes, Cibles et médicaments des infections et du cancer, IICiMed, EA 1155, 44000, Nantes, France
| | - Leslie Hesnard
- Université de Nantes, Inserm, CRCINA, 44000, Nantes, France
| | - Mikaël Croyal
- Université de Nantes, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, 44000, Nantes, France
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, 44000, Nantes, France
- CRNH-Ouest Mass Spectrometry Core Facility, 44000, Nantes, France
| | - Frédéric Pecorari
- P2R "Production de Protéines Recombinantes", Université de Nantes, CRCINA, SFR-Santé, INSERM, CNRS, CHU Nantes, Nantes, France
- Université de Nantes, Inserm, CRCINA, 44000, Nantes, France
| | - Nadine Gervois
- Université de Nantes, Inserm, CRCINA, 44000, Nantes, France.
- LabEx IGO «Immunotherapy, Graft, Oncology», Nantes, France.
| | - Karine Bernardeau
- P2R "Production de Protéines Recombinantes", Université de Nantes, CRCINA, SFR-Santé, INSERM, CNRS, CHU Nantes, Nantes, France.
- Université de Nantes, Inserm, CRCINA, 44000, Nantes, France.
| |
Collapse
|
14
|
Saso A, Kampmann B, Roetynck S. Vaccine-Induced Cellular Immunity against Bordetella pertussis: Harnessing Lessons from Animal and Human Studies to Improve Design and Testing of Novel Pertussis Vaccines. Vaccines (Basel) 2021; 9:877. [PMID: 34452002 PMCID: PMC8402596 DOI: 10.3390/vaccines9080877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Pertussis ('whooping cough') is a severe respiratory tract infection that primarily affects young children and unimmunised infants. Despite widespread vaccine coverage, it remains one of the least well-controlled vaccine-preventable diseases, with a recent resurgence even in highly vaccinated populations. Although the exact underlying reasons are still not clear, emerging evidence suggests that a key factor is the replacement of the whole-cell (wP) by the acellular pertussis (aP) vaccine, which is less reactogenic but may induce suboptimal and waning immunity. Differences between vaccines are hypothesised to be cell-mediated, with polarisation of Th1/Th2/Th17 responses determined by the composition of the pertussis vaccine given in infancy. Moreover, aP vaccines elicit strong antibody responses but fail to protect against nasal colonisation and/or transmission, in animal models, thereby potentially leading to inadequate herd immunity. Our review summarises current knowledge on vaccine-induced cellular immune responses, based on mucosal and systemic data collected within experimental animal and human vaccine studies. In addition, we describe key factors that may influence cell-mediated immunity and how antigen-specific responses are measured quantitatively and qualitatively, at both cellular and molecular levels. Finally, we discuss how we can harness this emerging knowledge and novel tools to inform the design and testing of the next generation of improved infant pertussis vaccines.
Collapse
Affiliation(s)
- Anja Saso
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Sophie Roetynck
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| |
Collapse
|
15
|
Yang J, Kim E, Lee JS, Poo H. A Murine CD8 + T Cell Epitope Identified in the Receptor-Binding Domain of the SARS-CoV-2 Spike Protein. Vaccines (Basel) 2021; 9:vaccines9060641. [PMID: 34208032 PMCID: PMC8230638 DOI: 10.3390/vaccines9060641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
The ongoing COVID-19 pandemic caused by SARS-CoV-2 has posed a devastating threat worldwide. The receptor-binding domain (RBD) of the spike protein is one of the most important antigens for SARS-CoV-2 vaccines, while the analysis of CD8 cytotoxic T lymphocyte activity in preclinical studies using mouse models is critical for evaluating vaccine efficacy. Here, we immunized C57BL/6 wild-type mice and transgenic mice expressing human angiotensin-converting enzyme 2 (ACE2) with the SARS-CoV-2 RBD protein to evaluate the IFN-γ-producing T cells in the splenocytes of the immunized mice using an overlapping peptide pool by an enzyme-linked immunospot assay and flow cytometry. We identified SARS-CoV-2 S395-404 as a major histocompatibility complex (MHC) class I-restricted epitope for the RBD-specific CD8 T cell responses in C57BL/6 mice.
Collapse
Affiliation(s)
- Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (J.Y.); (E.K.)
| | - Eunjin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (J.Y.); (E.K.)
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea;
| | - Jong-Soo Lee
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea;
| | - Haryoung Poo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (J.Y.); (E.K.)
- Correspondence: ; Tel.: +82-42-860-4157
| |
Collapse
|
16
|
Darwish M, Wichner S, Li J, Chang JC, Tam C, Franke Y, Li H, Chan P, Blanchette C. High-throughput identification of conditional MHCI ligands and scaled-up production of conditional MHCI complexes. Protein Sci 2021; 30:1169-1183. [PMID: 33840137 PMCID: PMC8138534 DOI: 10.1002/pro.4082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 01/28/2023]
Abstract
Despite the need to monitor the impact of Cancer Immunotherapy (CI)/Immuno-Oncology (IO) therapeutics on neoantigen-specific T-cell responses, very few clinical programs incorporate this aspect of immune monitoring due to the challenges in high-throughput (HTP) generation of Major Histocompatibility Complex Class I (MHCI) tetramers across a wide range of HLA alleles. This limitation was recently addressed through the development of MHCI complexes with peptides containing a nonnatural UV cleavable amino acid (conditional MHCI ligands) that enabled HTP peptide exchange upon UV exposure. Despite this advancement, the number of alleles with known conditional MHCI ligands is limited. We developed a novel workflow to enable identification and validation of conditional MHCI ligands across a range of HLA alleles. First, known peptide binders were screened via an enzyme-linked immunosorbent assay (ELISA) assay. Conditional MHCI ligands were designed using the highest-performing peptides and evaluated in the same ELISA assay. The top performers were then selected for scale-up production. Next-generation analytical techniques (LC/MS, SEC-MALS, and 2D LC/MS) were used to characterize the complex after refolding with the conditional MHCI ligands. Finally, we used 2D LC/MS to evaluate peptide exchange with these scaled-up conditional MHCI complexes after UV exposure with validated peptide binders. Successful peptide exchange was observed for all conditional MHCI ligands upon UV exposure, validating our screening approach. This approach has the potential to be broadly applied and enable HTP generation of MHCI monomers and tetramers across a wider range of HLA alleles, which could be critical to enabling the use of MHCI tetramers to monitor neoantigen-specific T-cells in the clinic.
Collapse
Affiliation(s)
| | - Sara Wichner
- Protein ChemistryGenentech IncSan FranciscoCaliforniaUSA
| | - Jenny Li
- Biochemical and Cellular PharmacologyGenentech IncSan FranciscoCaliforniaUSA
| | | | - Christine Tam
- BioMolecular ResourcesGenentech IncSan FranciscoCaliforniaUSA
| | - Yvonne Franke
- BioMolecular ResourcesGenentech IncSan FranciscoCaliforniaUSA
| | - Hong Li
- Protein ChemistryGenentech IncSan FranciscoCaliforniaUSA
| | - Pamela Chan
- Biochemical and Cellular PharmacologyGenentech IncSan FranciscoCaliforniaUSA
| | | |
Collapse
|
17
|
Moita D, Nunes-Cabaço H, Mendes AM, Prudêncio M. A guide to investigating immune responses elicited by whole-sporozoite pre-erythrocytic vaccines against malaria. FEBS J 2021; 289:3335-3359. [PMID: 33993649 DOI: 10.1111/febs.16016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/19/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022]
Abstract
In the last few decades, considerable efforts have been made toward the development of efficient vaccines against malaria. Whole-sporozoite (Wsp) vaccines, which induce efficient immune responses against the pre-erythrocytic (PE) stages (sporozoites and liver forms) of Plasmodium parasites, the causative agents of malaria, are among the most promising immunization strategies tested until present. Several Wsp PE vaccination approaches are currently under evaluation in the clinic, including radiation- or genetically-attenuated Plasmodium sporozoites, live parasites combined with chemoprophylaxis, or genetically modified rodent Plasmodium parasites. In addition to the assessment of their protective efficacy, clinical trials of Wsp PE vaccine candidates inevitably involve the thorough investigation of the immune responses elicited by vaccination, as well as the identification of correlates of protection. Here, we review the main methodologies employed to dissect the humoral and cellular immune responses observed in the context of Wsp PE vaccine clinical trials and discuss future strategies to further deepen the knowledge generated by these studies, providing a toolbox for the in-depth analysis of vaccine-induced immunogenicity.
Collapse
Affiliation(s)
- Diana Moita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - António M Mendes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| |
Collapse
|
18
|
Eddenden A, Kitova EN, Klassen JS, Nitz M. An Inactive Dispersin B Probe for Monitoring PNAG Production in Biofilm Formation. ACS Chem Biol 2020; 15:1204-1211. [PMID: 31917539 DOI: 10.1021/acschembio.9b00907] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The bacterial exopolysaccharide poly-β-1,6-N-acetylglucosamine is a major extracellular matrix component in biofilms of both Gram-positive and Gram-negative organisms. We have leveraged the specificity of the biofilm-dispersing glycoside hydrolase Dispersin B (DspB) to generate a probe (Dispersin B PNAG probe, DiPP) for monitoring PNAG production and localization during biofilm formation. Mutation of the active site of Dispersin B gave DiPP, which was an effective probe despite its low affinity for PNAG oligosaccharides (KD ∼ 1-10 mM). Imaging of PNAG-dependent and -independent biofilms stained with a fluorescent-protein fusion of DiPP (GFP-DiPP) demonstrated the specificity of the probe for the structure of PNAG on both single-cell and biofilm levels, indicating a high local concentration of PNAG at the bacterial cell surface. Through quantitative bacterial cell binding assays and confocal microscopy analysis using GFP-DiPP, discrete areas of local high concentrations of PNAG were detected on the surface of early log phase cells. These distinct areas were seen to grow, slough from cells, and accumulate in interbacterial regions over the course of several cell divisions, showing the development of a PNAG-dependent biofilm. A potential helical distribution of staining was also noted, suggesting some degree of organization of PNAG production at the cell surface prior to cell aggregation. Together, these experiments shed light on the early stages of PNAG-dependent biofilm formation and demonstrate the value of a low-affinity-high-specificity probe for monitoring the production of bacterial exopolysaccharides.
Collapse
Affiliation(s)
- Alexander Eddenden
- Department of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario, Canada M5S 3H6
| | - Elena N. Kitova
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr. Edmonton, Alberta, Canada T6G 2G2
| | - John S. Klassen
- Alberta Glycomics Centre and Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr. Edmonton, Alberta, Canada T6G 2G2
| | - Mark Nitz
- Department of Chemistry, University of Toronto, 80 St. George St, Toronto, Ontario, Canada M5S 3H6
| |
Collapse
|
19
|
Yuan Y, Brouchon J, Calvo-Calle JM, Xia J, Sun L, Zhang X, Clayton KL, Ye F, Weitz DA, Heyman JA. Droplet encapsulation improves accuracy of immune cell cytokine capture assays. LAB ON A CHIP 2020; 20:1513-1520. [PMID: 32242586 PMCID: PMC7313394 DOI: 10.1039/c9lc01261c] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Quantification of cell-secreted molecules, e.g., cytokines, is fundamental to the characterization of immune responses. Cytokine capture assays that use engineered antibodies to anchor the secreted molecules to the secreting cells are widely used to characterize immune responses because they allow both sensitive identification and recovery of viable responding cells. However, if the cytokines diffuse away from the secreting cells, non-secreting cells will also be identified as responding cells. Here we encapsulate immune cells in microfluidic droplets and perform in-droplet cytokine capture assays to limit the diffusion of the secreted cytokines. We use microfluidic devices to rapidly encapsulate single natural killer NK-92 MI cells and their target K562 cells into microfluidic droplets. We perform in-droplet IFN-γ capture assays and demonstrate that NK-92 MI cells recognize target cells within droplets and become activated to secrete IFN-γ. Droplet encapsulation prevents diffusion of secreted products to neighboring cells and dramatically reduces both false positives and false negatives, relative to assays performed without droplets. In a sample containing 1% true positives, encapsulation reduces, from 94% to 2%, the number of true-positive cells appearing as negatives; in a sample containing 50% true positives, the number of non-stimulated cells appearing as positives is reduced from 98% to 1%. After cells are released from the droplets, secreted cytokine remains captured onto secreting immune cells, enabling FACS-isolation of populations highly enriched for activated effector immune cells. Droplet encapsulation can be used to reduce background and improve detection of any single-cell secretion assay.
Collapse
Affiliation(s)
- Yuan Yuan
- Beijing National Laboratory for Condensed Matter Physics and CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Subramaniam KS, Lant S, Goodwin L, Grifoni A, Weiskopf D, Turtle L. Two Is Better Than One: Evidence for T-Cell Cross-Protection Between Dengue and Zika and Implications on Vaccine Design. Front Immunol 2020; 11:517. [PMID: 32269575 PMCID: PMC7109261 DOI: 10.3389/fimmu.2020.00517] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV, family Flaviviridae, genus Flavivirus) exists as four distinct serotypes. Generally, immunity after infection with one serotype is protective and lifelong, though exceptions have been described. However, secondary infection with a different serotype can result in more severe disease for a minority of patients. Host responses to the first DENV infection involve the development of both cross-reactive antibody and T cell responses, which, depending upon their precise balance, may mediate protection or enhance disease upon secondary infection with a different serotype. Abundant evidence now exists that responses elicited by DENV infection can cross-react with other members of the genus Flavivirus, particularly Zika virus (ZIKV). Cohort studies have shown that prior DENV immunity is associated with protection against Zika. Cross-reactive antibody responses may enhance infection with flaviviruses, which likely accounts for the cases of severe disease seen during secondary DENV infections. Data for T cell responses are contradictory, and even though cross-reactive T cell responses exist, their clinical significance is uncertain. Recent mouse experiments, however, show that cross-reactive T cells are capable of mediating protection against ZIKV. In this review, we summarize and discuss the evidence that T cell responses may, at least in part, explain the cross-protection seen against ZIKV from DENV infection, and that T cell antigens should therefore be included in putative Zika vaccines.
Collapse
Affiliation(s)
- Krishanthi S Subramaniam
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Suzannah Lant
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Lynsey Goodwin
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom.,Tropical and Infectious Disease Unit, Liverpool University Hospitals, Liverpool, United Kingdom
| |
Collapse
|
21
|
Haj AK, Breitbach ME, Baker DA, Mohns MS, Moreno GK, Wilson NA, Lyamichev V, Patel J, Weisgrau KL, Dudley DM, O'Connor DH. High-Throughput Identification of MHC Class I Binding Peptides Using an Ultradense Peptide Array. THE JOURNAL OF IMMUNOLOGY 2020; 204:1689-1696. [PMID: 32060132 DOI: 10.4049/jimmunol.1900889] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/04/2020] [Indexed: 01/02/2023]
Abstract
Rational vaccine development and evaluation requires identifying and measuring the magnitude of epitope-specific CD8 T cell responses. However, conventional CD8 T cell epitope discovery methods are labor intensive and do not scale well. In this study, we accelerate this process by using an ultradense peptide array as a high-throughput tool for screening peptides to identify putative novel epitopes. In a single experiment, we directly assess the binding of four common Indian rhesus macaque MHC class I molecules (Mamu-A1*001, -A1*002, -B*008, and -B*017) to ∼61,000 8-mer, 9-mer, and 10-mer peptides derived from the full proteomes of 82 SIV and simian HIV isolates. Many epitope-specific CD8 T cell responses restricted by these four MHC molecules have already been identified in SIVmac239, providing an ideal dataset for validating the array; up to 64% of these known epitopes are found in the top 192 SIVmac239 peptides with the most intense MHC binding signals in our experiment. To assess whether the peptide array identified putative novel CD8 T cell epitopes, we validated the method by IFN-γ ELISPOT assay and found three novel peptides that induced CD8 T cell responses in at least two Mamu-A1*001-positive animals; two of these were validated by ex vivo tetramer staining. This high-throughput identification of peptides that bind class I MHC will enable more efficient CD8 T cell response profiling for vaccine development, particularly for pathogens with complex proteomes for which few epitope-specific responses have been defined.
Collapse
Affiliation(s)
- Amelia K Haj
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Meghan E Breitbach
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - David A Baker
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Mariel S Mohns
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Gage K Moreno
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - Nancy A Wilson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | | | | | - Kim L Weisgrau
- Wisconsin National Primate Research Center, Madison, WI 53715
| | - Dawn M Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705; .,Wisconsin National Primate Research Center, Madison, WI 53715
| |
Collapse
|
22
|
Pastore G, Carraro M, Pettini E, Nolfi E, Medaglini D, Ciabattini A. Optimized Protocol for the Detection of Multifunctional Epitope-Specific CD4 + T Cells Combining MHC-II Tetramer and Intracellular Cytokine Staining Technologies. Front Immunol 2019; 10:2304. [PMID: 31649661 PMCID: PMC6794358 DOI: 10.3389/fimmu.2019.02304] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
Analysis of multifunctional CD4+ T cells is fundamental for characterizing the immune responses to vaccination or infection. Major histocompatibility complex (MHC)/peptide tetramers represent a powerful technology for the detection of antigen-specific T cells by specific binding to their T-cell receptor, and their combination with functional assays is fundamental for characterizing the antigen-specific immune response. Here we optimized a protocol for the detection of multiple intracellular cytokines within epitope-specific CD4+ T cells identified by the MHC class II tetramer technology. The optimal procedure for assessing the functional activity of tetramer-binding CD4+ T cells was based on the simultaneous intracellular staining with both MHC tetramers and cytokine-specific antibodies upon in vitro restimulation of cells with the vaccine antigen. The protocol was selected among procedures that differently combine the steps of cellular restimulation and tetramer staining with intracellular cytokine labeling. This method can be applied to better understand the complex functional profile of CD4+ T-cell responses upon vaccination or infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
23
|
FACS isolation of low percentage human antigen-specific CD8 + T cells based on activation-induced CD3 and CD8 downregulation. J Immunol Methods 2019; 472:35-43. [PMID: 31201792 DOI: 10.1016/j.jim.2019.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 11/21/2022]
Abstract
As T cell activation leads to downregulation of T cell receptor (TCR) and coreceptor CD8, we developed a novel FACS-based sorting method to enrich activated antigen-specific CD8+ T cells. Using multiple established or low percentage T cell cultures, with either single antigen specificity or multiple influenza A virus antigen specificities, we have optimized the sorting method for T cell activation time and stimulating antigen dose. We have also sorted various numbers of antigen-specific CD8+ T cells into 96-well plates to demonstrate these T cells are capable of expanding into nearly pure CD8+ T cell lines. Our approach has the advantage of sorting antigen-specific T cells without knowing their specific antigenic epitopes or restricting HLA. We believe this method can be very helpful for successfully establishing CD8+ T cell lines for various purpose, including immunotherapy.
Collapse
|
24
|
Sung PS, Park DJ, Kim JH, Han JW, Lee EB, Lee GW, Nam HC, Jang JW, Bae SH, Choi JY, Shin EC, Park SH, Yoon SK. Ex vivo Detection and Characterization of Hepatitis B Virus-Specific CD8 + T Cells in Patients Considered Immune Tolerant. Front Immunol 2019; 10:1319. [PMID: 31244857 PMCID: PMC6563765 DOI: 10.3389/fimmu.2019.01319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/23/2019] [Indexed: 12/21/2022] Open
Abstract
In this study, we aimed to detect and characterize ex vivo virus-specific CD8+ T cells in patients with immune-tolerant hepatitis B virus (HBV) infection. We investigated a Korean chronic hepatitis B cohort composed of 15 patients in the immune-tolerant phase, 17 in the immune-active phase, and 13 under antiviral treatment. We performed enzyme-linked immunospot (ELISpot) assays ex vivo and intracellular cytokine staining after in vitro culture. We also performed ex vivo multimer staining assays and examined the expression of programmed death-1 (PD-1) and CD127 in pentamer-positive cells. Ex vivo ELISpot revealed that HBV-specific T cell function was weaker in immune-tolerant patients than in those under antiviral treatment. In vitro culture of peripheral blood mononuclear cells for 10 days revealed that HBV-specific CD8+ T cells produced interferon-γ in some immune-tolerant patients. We detected HBV-specific CD8+ T cells ex vivo (using the HBV core18-27 pentamer) in patients from all three groups. The PD-1+ subset of pentamer+ CD8+ T cells was smaller ex vivo in the immune-tolerant phase than in the immune-active phase or under antiviral treatment. Interestingly, the proportion of PD-1+ CD8+ T cells in HBV-specific CD8+ T cells correlated with patient age when all enrolled patients were analyzed. Overall, HBV-specific CD8+ T cells are present in patients considered as immune-tolerant, although their ex vivo functionality is significantly weaker than that in patients under antiviral treatment (P < 0.05). Despite the high viral load, the proportion of PD-1 expression in HBV-specific CD8+ T cells is lower in the immune-tolerant phase than in other phases. Our results indicate appropriate stimulation may enhance the effector function of HBV-specific CD8+ T cells in patients considered as being in the immune-tolerant phase.
Collapse
Affiliation(s)
- Pil Soo Sung
- Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Dong Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jung-Hee Kim
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Won Han
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, South Korea
| | - Eun Byul Lee
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Gil Won Lee
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hee Chul Nam
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Jang
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Si Hyun Bae
- Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea.,Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong Young Choi
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, South Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, South Korea
| | - Seung Kew Yoon
- Department of Biomedicine & Health Sciences, The Catholic University Liver Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
25
|
Xu A, Zhang L, Yuan J, Babikr F, Freywald A, Chibbar R, Moser M, Zhang W, Zhang B, Fu Z, Xiang J. TLR9 agonist enhances radiofrequency ablation-induced CTL responses, leading to the potent inhibition of primary tumor growth and lung metastasis. Cell Mol Immunol 2018; 16:820-832. [PMID: 30467420 DOI: 10.1038/s41423-018-0184-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/25/2018] [Indexed: 12/11/2022] Open
Abstract
Radiofrequency ablation (RFA) is the most common approach to thermal ablation for cancer therapy. Unfortunately, its efficacy is limited by incomplete ablation, and further optimization of RFA is required. Here, we demonstrate that incubation at 65 °C triggers more EG7 tumor cell death by necrosis than treatment at 45 °C, and the 65 °C-treated cells are more effective at inducing antigen-specific CD8+ cytotoxic T lymphocyte (CTL) responses after injection in mice than the 45 °C-treated ones. Dendritic cells (DCs) that phagocytose 65 °C-treated EG7 cells become mature with upregulated MHCII and CD80 expression and are capable of efficiently inducing effector CTLs in mouse tumor models. RFA (65 °C) therapy of EG7 tumors induces large areas of tumor necrosis and stimulates CTL responses. This leads to complete regression of small (~100 mm3) tumors but fails to suppress the growth of larger (~350 mm3) tumors. The administration of the Toll-like receptor-9 (TLR9) agonist unmethylated cytosine-phosphorothioate-guanine oligonucleotide (CpG) to DCs phagocytosing 65 °C-treated EG7 cells enhances the expression of MHCII and CD40 on DCs as well as DC-induced stimulation of CTL responses. Importantly, the intratumoral administration of CpG following RFA also increases the frequencies of tumor-associated immunogenic CD11b-CD11c+CD103+ DC2 and CD11b+F4/80+MHCII+ M1 macrophages and increases CD4+ and CD8+ T-cell tumor infiltration, leading to enhanced CD4+ T cell-dependent CTL responses and potent inhibition of primary RFA-treated or distant untreated tumor growth as well as tumor lung metastasis in mice bearing larger tumors. Overall, our data indicate that CpG administration, which enhances RFA-induced CTL responses and ultimately potentiates the inhibition of primary tumor growth and lung metastasis, is a promising strategy for improving RFA treatment, which may assist in optimizing this important cancer therapy.
Collapse
Affiliation(s)
- Aizhang Xu
- Cancer Research, Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Oncology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lifeng Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingying Yuan
- Cancer Research, Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Oncology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Fatma Babikr
- Cancer Research, Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon, SK, Canada.,Department of Oncology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Andrew Freywald
- Department of Pathology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Rajni Chibbar
- Department of Pathology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Michael Moser
- Department of Surgery, University of Saskatchewan, Saskatoon, SK, Canada
| | - Wenjun Zhang
- Department of Bioengineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Bing Zhang
- Biomedical Science and Technology Research Center, School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
| | - Zhaoying Fu
- Department of Immunology, College of Medicine, Yian-An University, Yian-An, China
| | - Jim Xiang
- Cancer Research, Saskatchewan Cancer Agency, University of Saskatchewan, Saskatoon, SK, Canada. .,Department of Oncology, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
26
|
Sorini C, Cardoso RF, Gagliani N, Villablanca EJ. Commensal Bacteria-Specific CD4 + T Cell Responses in Health and Disease. Front Immunol 2018; 9:2667. [PMID: 30524431 PMCID: PMC6256970 DOI: 10.3389/fimmu.2018.02667] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Over the course of evolution, mammalian body surfaces have adapted their complex immune system to allow a harmless coexistence with the commensal microbiota. The adaptive immune response, in particular CD4+ T cell-mediated, is crucial to maintain intestinal immune homeostasis by discriminating between harmless (e.g., dietary compounds and intestinal microbes) and harmful stimuli (e.g., pathogens). To tolerate food molecules and microbial components, CD4+ T cells establish a finely tuned crosstalk with the environment whereas breakdown of these mechanisms might lead to chronic disease associated with mucosal barriers and beyond. How commensal-specific immune responses are regulated and how these molecular and cellular mechanisms can be manipulated to treat chronic disorders is yet poorly understood. In this review, we discuss current knowledge of the regulation of commensal bacteria-specific CD4+ T cells. We place particular focus on the key role of commensal-specific CD4+ T cells in maintaining tolerance while efficiently eradicating local and systemic infections, with a focus on factors that trigger their aberrant activation.
Collapse
Affiliation(s)
- Chiara Sorini
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Rebeca F. Cardoso
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Nicola Gagliani
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eduardo J. Villablanca
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
27
|
A novel and simple method to produce large amounts of recombinant soluble peptide/major histocompatibility complex monomers for analysis of antigen-specific human T cell receptors. N Biotechnol 2018; 49:169-177. [PMID: 30465909 DOI: 10.1016/j.nbt.2018.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 11/14/2018] [Accepted: 11/18/2018] [Indexed: 11/24/2022]
Abstract
Soluble peptide/major histocompatibility complex (p/MHC) tetramers that directly bind to T cell receptors (TCRs) allow the direct quantification, phenotypic characterization and isolation of antigen-specific T cells. Conventionally, soluble p/MHC tetramers have been produced using Escherichia coli, but this method requires refolding of the recombinant proteins. Here, a novel and technically simple method that does not require protein refolding in vitro has been developed for the high-throughput generation of soluble and functional p/MHC-single chain trimer (SCT) monomers and tetramers in a mammalian cell system. The p/MHC-SCT tetramers generated by this method bound to the corresponding antigen-specific TCRs. Moreover, the immobilized p/MHC-SCT monomers effectively activated antigen-specific T cell lines as well as primary T cells in an antigen-specific manner. This technique provides a robust improvement in the technology, such that recombinant soluble p/MHC monomers and tetramers can be produced more readily and which enables their use in analysis of antigen-specific T cells in basic and clinical studies.
Collapse
|
28
|
Place DE, Williamson DR, Yuzefpolskiy Y, Katkere B, Sarkar S, Kalia V, Kirimanjeswara GS. Development of a novel Francisella tularensis Live Vaccine Strain expressing ovalbumin provides insight into antigen-specific CD8+ T cell responses. PLoS One 2017; 12:e0190384. [PMID: 29284034 PMCID: PMC5746256 DOI: 10.1371/journal.pone.0190384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 12/13/2017] [Indexed: 01/19/2023] Open
Abstract
Progress towards a safe and effective vaccine for the prevention of tularemia has been hindered by a lack of knowledge regarding the correlates of protective adaptive immunity and a lack of tools to generate this knowledge. CD8+ T cells are essential for protective immunity against virulent strains of Francisella tularensis, but to-date, it has not been possible to study these cells in an antigen-specific manner. Here, we report the development of a tool for expression of the model antigen ovalbumin (OVA) in F. tularensis, which allows for the study of CD8+ T cell responses to the bacterium. We demonstrate that in response to intranasal infection with the F. tularensis Live Vaccine Strain, adoptively transferred OVA-specific CD8+ T cells expand after the first week and produce IFN-γ but not IL-17. Effector and central memory subsets develop with disparate kinetics in the lungs, draining lymph node and spleen. Notably, OVA-specific cells are poorly retained in the lungs after clearance of infection. We also show that intranasal vaccination leads to more antigen-specific CD8+ T cells in the lung-draining lymph node compared to scarification vaccination, but that an intranasal booster overcomes this difference. Together, our data show that this novel tool can be used to study multiple aspects of the CD8+ T cell response to F. tularensis. Use of this tool will enhance our understanding of immunity to this deadly pathogen.
Collapse
Affiliation(s)
- David E. Place
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - David R. Williamson
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Yevgeniy Yuzefpolskiy
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Bhuvana Katkere
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Surojit Sarkar
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Vandana Kalia
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Girish S. Kirimanjeswara
- Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
29
|
Leitman EM, Palmer CD, Buus S, Chen F, Riddell L, Sims S, Klenerman P, Sáez-Cirión A, Walker BD, Hess PR, Altfeld M, Matthews PC, Goulder PJR. Saporin-conjugated tetramers identify efficacious anti-HIV CD8+ T-cell specificities. PLoS One 2017; 12:e0184496. [PMID: 29020090 PMCID: PMC5636067 DOI: 10.1371/journal.pone.0184496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/24/2017] [Indexed: 12/25/2022] Open
Abstract
Antigen-specific T-cells are highly variable, spanning potent antiviral efficacy and damaging auto-reactivity. In virus infections, identifying the most efficacious responses is critical to vaccine design. However, current methods depend on indirect measures or on ex vivo expanded CTL clones. We here describe a novel application of cytotoxic saporin-conjugated tetramers to kill antigen-specific T-cells without significant off-target effects. The relative efficacy of distinct antiviral CD8+ T-cell specificity can be directly assessed via antigen-specific CD8+ T-cell depletion. The utility of these reagents is demonstrated here in identifying the CD8+ T-cell specificity most effective in preventing HIV progression in HIV-infected HLA-B*27-positive immune controllers.
Collapse
Affiliation(s)
- Ellen M. Leitman
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Christine D. Palmer
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Søren Buus
- Laboratory of Experimental Immunology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fabian Chen
- Department of Sexual Health, Royal Berkshire Hospital, Reading, United Kingdom
| | - Lynn Riddell
- Integrated Sexual Health Services, Northamptonshire Healthcare NHS Trust, Northampton, United Kingdom
| | - Stuart Sims
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Paul Klenerman
- NIHR Biomedical Research Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Asier Sáez-Cirión
- Institut Pasteur, Unité HIV, Inflammation et Persistance, Paris, France
| | - Bruce D. Walker
- Harvard Medical School, Boston, Massachusetts, United States of America
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Paul R. Hess
- Immunology Program, Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, United States of America
| | - Marcus Altfeld
- Harvard Medical School, Boston, Massachusetts, United States of America
- Virus Immunology Unit, Heinrich-Pette-Institut, Hamburg, Germany
| | - Philippa C. Matthews
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Infectious Diseases and Microbiology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Philip J. R. Goulder
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- * E-mail:
| |
Collapse
|
30
|
Zhu J, Huang Y, Su J, He J, Yu Y, Zhao Y, Lu X. Quantum dot/pMHC multimers vs. phycoerythrin/pMHC tetramers for identification of HLA-A*0201-restricted pHBV core antigen18-27-specific T cells. Mol Med Rep 2017; 16:8605-8612. [PMID: 28765875 PMCID: PMC5779913 DOI: 10.3892/mmr.2017.7126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/01/2017] [Indexed: 11/06/2022] Open
Abstract
Detection of human leukocyte antigens-A2-restricted p-hepatitis B virus (HBV) core antigen‑specific cytotoxic T lymphocytes (CTLs) is important in the study of HBV immunopathogenesis and vaccine design. Currently, major histocompatibility complex (MHC) class I/peptide‑(p) MHCI tetramers are considered the optimal tools to detect antigen‑specific CTLs. However, the MHC‑tetramer technique also has certain drawbacks and is under continuous development. The quantum dot (QD) bioconjugates nanotechnology with its unique inorganic‑biological properties has been developing fast. However, QD/pMHC multimers have seldom been used for the identification of the C18‑27 epitope, which is important in HBV infection. QD/pMHC multimers were synthesized by metal‑affinity coordination and an avidin‑biotin system. In the present study they were characterized by transmission electron microscopy, dynamic light scattering and fluorescence spectrophotometry. C18‑27‑specific CTLs were obtained by ex vivo expansion of CD8+ T cells. Cultured CTLs were tested for the secretion level of interferon (IFN)‑γ by ELISA and for cytotoxicity by lactate dehydrogenase release assay. Then, the performance of phycoerythrin (PE)/pMHC tetramers and QD/pMHC multimers were compared by flow cytometry. The synthesized QD/pMHC multimers dispersed well and their emission spectrum exhibited only slight differences compared with original QDs. C18‑27‑specific CTLs not only secreted IFN‑γ but also effectively targeted T2 cells pulsed with peptide C18‑27. The frequencies of C18‑27‑specific CTLs determined by QD/pMHC multimers were higher compared with PE/pMHC tetramers. The present results suggested that QD/pMHC multimers may be able to characterize greater numbers of C18‑27‑specific CTLs with increased sensitivity compared to conventional strategies.
Collapse
Affiliation(s)
- Jianmeng Zhu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yong Huang
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing Su
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jian He
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yating Yu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoling Lu
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
31
|
Svitek N, Taracha ELN, Saya R, Awino E, Nene V, Steinaa L. Analysis of the Cellular Immune Responses to Vaccines. Methods Mol Biol 2016; 1349:247-262. [PMID: 26458841 DOI: 10.1007/978-1-4939-3008-1_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Flow cytometry, enzyme-linked immunospot (ELISpot) and cellular cytotoxicity assays are powerful tools for studying the cellular immune response towards intracellular pathogens and vaccines in livestock species. Lymphocytes from immunized animals can be purified using Ficoll-Paque density gradient centrifugation and evaluated for their antigen specificity or reactivity towards a vaccine. Here, we describe staining of bovine lymphocytes with peptide (p)-MHC class I tetramers and antibodies specific towards cellular activation markers for evaluation by multiparametric flow cytometry, as well as interferon (IFN)-γ ELISpot and cytotoxicity using chromium ((51)Cr) release assays. A small component on the use of immunoinformatics for fine-tuning the identification of a minimal CTL epitope is included.
Collapse
Affiliation(s)
- Nicholas Svitek
- Vaccine Biosciences, International Livestock Research Institute, Att: Lucilla Steinaa, Vaccine Biosciences, 30709, 00100, Nairobi, Kenya
| | - Evans L N Taracha
- Vaccine Biosciences, International Livestock Research Institute, Att: Lucilla Steinaa, Vaccine Biosciences, 30709, 00100, Nairobi, Kenya
| | - Rosemary Saya
- Vaccine Biosciences, International Livestock Research Institute, Att: Lucilla Steinaa, Vaccine Biosciences, 30709, 00100, Nairobi, Kenya
| | - Elias Awino
- Vaccine Biosciences, International Livestock Research Institute, Att: Lucilla Steinaa, Vaccine Biosciences, 30709, 00100, Nairobi, Kenya
| | - Vishvanath Nene
- Vaccine Biosciences, International Livestock Research Institute, Att: Lucilla Steinaa, Vaccine Biosciences, 30709, 00100, Nairobi, Kenya
| | - Lucilla Steinaa
- Vaccine Biosciences, International Livestock Research Institute, Att: Lucilla Steinaa, Vaccine Biosciences, 30709, 00100, Nairobi, Kenya.
| |
Collapse
|
32
|
Dutta DK, Rhodes K, Wood SC. In silico prediction of Ebola Zaire GP(1,2) immuno-dominant epitopes for the Balb/c mouse. BMC Immunol 2015; 16:59. [PMID: 26445317 PMCID: PMC4596288 DOI: 10.1186/s12865-015-0126-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/01/2015] [Indexed: 12/04/2022] Open
Abstract
Background Ebola is a Filovirus (FV) that induces a highly communicable and deadly hemorrhagic fever. Currently, there are no approved vaccines to treat FV infections. Protection from FV infection requires cell mediated and humoral immunity. Glycoprotein GP1,2 Fc Zaire, a recombinant FV human Fc fusion protein, has been shown to confer protection against mouse adapted Zaire Ebola virus. The present studies are focused upon identifying immunodominant epitopes using in silico methods and developing tetramers as a diagnostic reagent to detect cell mediated immune responses to GP1,2 Fc. Methods The GP1,2 Ebola Zaire sequence from the 1976 outbreak was analyzed by both BIMAS and SYFPEITHI algorithms to identify potential immuno-dominant epitopes. Several peptides were synthesized and screened in flow-based MHC stability studies. Three candidate peptides, P8, P9 and P10, were identified and, following immunization in Balb/c mice, all three peptides induced IFN-γ as detected by ELISpot and intracellular staining. Results Significantly, P8, P9 and P10 generated robust cytotoxic T-cell responses (CTL) as determined by a flow cytometry-based Caspase assay. Antigen specific cells were also detected, using tetramers. Both P9 and P10 have sequence homology with highly conserved regions of several strains of FV. Conclusions In sum, three immunodominant sequences of the Ebola GP1,2 have been identified using in silico methods that may confer protection against mouse adapted Ebola Zaire. The development of tetramer reagents will provide unique insight into the potency and durability of medical countermeasure vaccines for known bioterrorism threat agents in preclinical models.
Collapse
Affiliation(s)
- Debargh K Dutta
- Division of Biology, Chemistry and Materials Science, Center for Devices and Radiological Health, FDA, Silver Spring, MD, 20993, USA. .,Department of Medicine, USUHS, 4301 Jones Bridge road, Bethesda, MD, 20814, USA.
| | - Kelly Rhodes
- Division of Biology, Chemistry and Materials Science, Center for Devices and Radiological Health, FDA, Silver Spring, MD, 20993, USA. .,University of Maryland, College Park, MD, 20742, USA.
| | - Steven C Wood
- Division of Biology, Chemistry and Materials Science, Center for Devices and Radiological Health, FDA, Silver Spring, MD, 20993, USA.
| |
Collapse
|
33
|
Phetsouphanh C, Zaunders JJ, Kelleher AD. Detecting Antigen-Specific T Cell Responses: From Bulk Populations to Single Cells. Int J Mol Sci 2015; 16:18878-93. [PMID: 26274954 PMCID: PMC4581277 DOI: 10.3390/ijms160818878] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/18/2022] Open
Abstract
A new generation of sensitive T cell-based assays facilitates the direct quantitation and characterization of antigen-specific T cell responses. Single-cell analyses have focused on measuring the quality and breadth of a response. Accumulating data from these studies demonstrate that there is considerable, previously-unrecognized, heterogeneity. Standard assays, such as the ICS, are often insufficient for characterization of rare subsets of cells. Enhanced flow cytometry with imaging capabilities enables the determination of cell morphology, as well as the spatial localization of the protein molecules within a single cell. Advances in both microfluidics and digital PCR have improved the efficiency of single-cell sorting and allowed multiplexed gene detection at the single-cell level. Delving further into the transcriptome of single-cells using RNA-seq is likely to reveal the fine-specificity of cellular events such as alternative splicing (i.e., splice variants) and allele-specific expression, and will also define the roles of new genes. Finally, detailed analysis of clonally related antigen-specific T cells using single-cell TCR RNA-seq will provide information on pathways of differentiation of memory T cells. With these state of the art technologies the transcriptomics and genomics of Ag-specific T cells can be more definitively elucidated.
Collapse
Affiliation(s)
| | - John James Zaunders
- Kirby Institute, University of New South Wales, 2031 Sydney, Australia.
- Centre for Applied Medical Research, St. Vincent's Hospital, 2010 Sydney, Australia.
| | - Anthony Dominic Kelleher
- Kirby Institute, University of New South Wales, 2031 Sydney, Australia.
- Centre for Applied Medical Research, St. Vincent's Hospital, 2010 Sydney, Australia.
| |
Collapse
|
34
|
Abstract
The binding between biotin and streptavidin or avidin is one of the strongest known non-covalent biological interactions. The (strept)avidin-biotin interaction has been widely used for decades in biological research and biotechnology. Therefore labeling of purified proteins by biotin is a powerful way to achieve protein capture, immobilization, and functionalization, as well as multimerizing or bridging molecules. Chemical biotinylation often generates heterogeneous products, which may have impaired function. Enzymatic biotinylation with E. coli biotin ligase (BirA) is highly specific in covalently attaching biotin to the 15 amino acid AviTag peptide, giving a homogeneous product with high yield. AviTag can conveniently be added genetically at the N-terminus, C-terminus, or in exposed loops of a target protein. We describe here procedures for AviTag insertion by inverse PCR, purification of BirA fused to glutathione-S-transferase (GST-BirA) from E. coli, BirA biotinylation of purified protein, and gel-shift analysis by SDS-PAGE to quantify the extent of biotinylation.
Collapse
Affiliation(s)
- Michael Fairhead
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | | |
Collapse
|
35
|
Reguzova AY, Karpenko LI, Mechetina LV, Belyakov IM. Peptide-MHC multimer-based monitoring of CD8 T-cells in HIV-1 infection and AIDS vaccine development. Expert Rev Vaccines 2014; 14:69-84. [PMID: 25373312 DOI: 10.1586/14760584.2015.962520] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of MHC multimers allows precise and direct detecting and analyzing of antigen-specific T-cell populations and provides new opportunities to characterize T-cell responses in humans and animals. MHC-multimers enable us to enumerate specific T-cells targeting to viral, tumor and vaccine antigens with exceptional sensitivity and specificity. In the field of HIV/SIV immunology, this technique provides valuable information about the frequencies of HIV- and SIV-specific CD8(+) cytotoxic T lymphocytes (CTLs) in different tissues and sites of infection, AIDS progression, and pathogenesis. Peptide-MHC multimer technology remains a very sensitive tool in detecting virus-specific T -cells for evaluation of the immunogenicity of vaccines against HIV-1 in preclinical trials. Moreover, it helps to understand how immune responses are formed following vaccination in the dynamics from priming point until T-cell memory is matured. Here we review a diversity of peptide-MHC class I multimer applications for fundamental immunological studies in different aspects of HIV/SIV infection and vaccine development.
Collapse
Affiliation(s)
- Alena Y Reguzova
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk region, 630559, Russia
| | | | | | | |
Collapse
|
36
|
Podetz-Pedersen KM, Vezys V, Somia NV, Russell SJ, McIvor RS. Cellular immune response against firefly luciferase after sleeping beauty-mediated gene transfer in vivo. Hum Gene Ther 2014; 25:955-65. [PMID: 25093708 PMCID: PMC4251089 DOI: 10.1089/hum.2014.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 08/01/2014] [Indexed: 12/12/2022] Open
Abstract
The Sleeping Beauty (SB) transposon system has been shown to mediate new gene sequence integration resulting in long-term expression. Here the effectiveness of hyperactive SB100X transposase was tested, and we found that hydrodynamic co-delivery of a firefly luciferase transposon (pT2/CaL) along with SB100X transposase (pCMV-SB100X) resulted in remarkably sustained, high levels of luciferase expression. However, after 4 weeks there was a rapid, animal-by-animal loss of luciferase expression that was not observed in immunodeficient mice. We hypothesized that this sustained, high-level luciferase expression achieved using the SB100X transposase elicits an immune response in pT2/CaL co-administered mice, which was supported by the rapid loss of luciferase expression upon challenge of previously treated animals and in naive animals adoptively transferred with splenocytes from previously treated animals. Specificity of the immune response to luciferase was demonstrated by increased cytokine expression in splenocytes after exposure to luciferase peptide in parallel with MHC I-luciferase peptide tetramer binding. This anti-luciferase immune response observed following continuous, high-level luciferase expression in vivo clearly impacts its use as an in vivo reporter. As both an immunogen and an extremely sensitive reporter, luciferase is also a useful model system for the study of immune responses following in vivo gene transfer and expression.
Collapse
Affiliation(s)
- Kelly M. Podetz-Pedersen
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455
| | - Vaiva Vezys
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Nikunj V. Somia
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455
| | | | - R. Scott McIvor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
37
|
Svitek N, Hansen AM, Steinaa L, Saya R, Awino E, Nielsen M, Buus S, Nene V. Use of "one-pot, mix-and-read" peptide-MHC class I tetramers and predictive algorithms to improve detection of cytotoxic T lymphocyte responses in cattle. Vet Res 2014; 45:50. [PMID: 24775445 PMCID: PMC4018993 DOI: 10.1186/1297-9716-45-50] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 04/07/2014] [Indexed: 11/10/2022] Open
Abstract
Peptide-major histocompatibility complex (p-MHC) class I tetramer complexes have facilitated the early detection and functional characterisation of epitope specific CD8+ cytotoxic T lymphocytes (CTL). Here, we report on the generation of seven recombinant bovine leukocyte antigens (BoLA) and recombinant bovine β2-microglobulin from which p-MHC class I tetramers can be derived in ~48 h. We validated a set of p-MHC class I tetramers against a panel of CTL lines specific to seven epitopes on five different antigens of Theileria parva, a protozoan pathogen causing the lethal bovine disease East Coast fever. One of the p-MHC class I tetramers was tested in ex vivo assays and we detected T. parva specific CTL in peripheral blood of cattle at day 15-17 post-immunization with a live parasite vaccine. The algorithm NetMHCpan predicted alternative epitope sequences for some of the T. parva CTL epitopes. Using an ELISA assay to measure peptide-BoLA monomer formation and p-MHC class I tetramers of new specificity, we demonstrate that a predicted alternative epitope Tp229-37 rather than the previously reported Tp227-37 epitope is the correct Tp2 epitope presented by BoLA-6*04101. We also verified the prediction by NetMHCpan that the Tp587-95 epitope reported as BoLA-T5 restricted can also be presented by BoLA-1*02301, a molecule similar in sequence to BoLA-T5. In addition, Tp587-95 specific bovine CTL were simultaneously stained by Tp5-BoLA-1*02301 and Tp5-BoLA-T5 tetramers suggesting that one T cell receptor can bind to two different BoLA MHC class I molecules presenting the Tp587-95 epitope and that these BoLA molecules fall into a single functional supertype.
Collapse
Affiliation(s)
- Nicholas Svitek
- International Livestock Research Institute (ILRI), P,O, Box 30709, Nairobi 00100, Kenya.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Fairhead M, Krndija D, Lowe ED, Howarth M. Plug-and-play pairing via defined divalent streptavidins. J Mol Biol 2014; 426:199-214. [PMID: 24056174 PMCID: PMC4047826 DOI: 10.1016/j.jmb.2013.09.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/07/2013] [Accepted: 09/12/2013] [Indexed: 11/29/2022]
Abstract
Streptavidin is one of the most important hubs for molecular biology, either multimerizing biomolecules, bridging one molecule to another, or anchoring to a biotinylated surface/nanoparticle. Streptavidin has the advantage of rapid ultra-stable binding to biotin. However, the ability of streptavidin to bind four biotinylated molecules in a heterogeneous manner is often limiting. Here, we present an efficient approach to isolate streptavidin tetramers with two biotin-binding sites in a precise arrangement, cis or trans. We genetically modified specific subunits with negatively charged tags, refolded a mixture of monomers, and used ion-exchange chromatography to resolve tetramers according to the number and orientation of tags. We solved the crystal structures of cis-divalent streptavidin to 1.4Å resolution and trans-divalent streptavidin to 1.6Å resolution, validating the isolation strategy and explaining the behavior of the Dead streptavidin variant. cis- and trans-divalent streptavidins retained tetravalent streptavidin's high thermostability and low off-rate. These defined divalent streptavidins enabled us to uncover how streptavidin binding depends on the nature of the biotin ligand. Biotinylated DNA showed strong negative cooperativity of binding to cis-divalent but not trans-divalent streptavidin. A small biotinylated protein bound readily to cis and trans binding sites. We also solved the structure of trans-divalent streptavidin bound to biotin-4-fluorescein, showing how one ligand obstructs binding to an adjacent biotin-binding site. Using a hexaglutamate tag proved a more powerful way to isolate monovalent streptavidin, for ultra-stable labeling without undesired clustering. These forms of streptavidin allow this key hub to be used with a new level of precision, for homogeneous molecular assembly.
Collapse
Affiliation(s)
- Michael Fairhead
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Denis Krndija
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Ed D Lowe
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Mark Howarth
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
39
|
Tools and methods for identification and analysis of rare antigen-specific T lymphocytes. EXPERIENTIA SUPPLEMENTUM (2012) 2014; 104:73-88. [PMID: 24214619 DOI: 10.1007/978-3-0348-0726-5_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
T lymphocytes are essential as effector and memory cells for immune defense against infections and as regulatory T cells in the establishment and maintenance of immune tolerance. However, they are also involved in immune pathology being effectors in autoimmune and allergic diseases or suppressors of immunity in cancer, and they often cause problems in transplantation. Therefore, strategies are being developed that allow the in vivo amplification or isolation, in vitro expansion and genetic manipulation of beneficial T cells for adoptive cell therapies or for the tolerization, or elimination of pathogenic T cells. The major goal is to make use of the exquisite antigen specificity of T cells to develop targeted strategies and to develop techniques that allow for the identification and depletion or enrichment of very often rare antigen-specific naïve as well as effector and memory T cells. Such techniques are very useful for immune monitoring of T cell responses in diagnostics and vaccination and for the development of T cell-based assays for the replacement of animal testing in immunotoxicology to identify contact allergens and drugs that cause adverse reactions.
Collapse
|
40
|
Xu D, Prasad S, Miller SD. Inducing immune tolerance: a focus on Type 1 diabetes mellitus. ACTA ACUST UNITED AC 2013; 3:415-426. [PMID: 24505231 DOI: 10.2217/dmt.13.36] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Tolerogenic strategies that specifically target diabetogenic immune cells in the absence of complications of immunosuppression are the desired treatment for the prevention or even reversal of Type 1 diabetes (T1D). Antigen (Ag)-based therapies must not only suppress disease-initiating diabetogenic T cells that are already activated, but, more importantly, prevent activation of naive auto-Ag-specific T cells that may become autoreactive through epitope spreading as a result of Ag liberation from damaged islet cells. Therefore, identification of auto-Ags relevant to T1D initiation and progression is critical to the design of effective Ag-specific therapies. Animal models of T1D have been successfully employed to identify potential diabetogenic Ags, and have further facilitated translation of Ag-specific tolerance strategies into human clinical trials. In this review, we highlight important advances using animal models in Ag-specific T1D immunotherapies, and the application of the preclinical findings to human subjects. We provide an up-to-date overview of the strengths and weaknesses of various tolerance-inducing strategies, including infusion of soluble Ags/peptides by various routes of delivery, genetic vaccinations, cell- and inert particle-based tolerogenic approaches, and various other strategies that target distinct tolerance-inducing pathways.
Collapse
Affiliation(s)
- Dan Xu
- Department of Microbiology-Immunology & Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA
| | - Suchitra Prasad
- Department of Microbiology-Immunology & Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology & Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, 303 E Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|
41
|
Bordignon V, Cordiali-Fei P, Rinaldi M, Signori E, Cottarelli A, Zonfrillo M, Ensoli F, Rasi G, Fuggetta MP. Evaluation of antigen specific recognition and cell mediated cytotoxicity by a modified lysispot assay in a rat colon carcinoma model. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:9. [PMID: 22296726 PMCID: PMC3395825 DOI: 10.1186/1756-9966-31-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/01/2012] [Indexed: 12/29/2022]
Abstract
Background Antigen-specific CD8+ cytotoxic T lymphocytes represent potent effector cells of the adaptive immune response against viruses as well as tumours. Therefore assays capable at exploring the generation and function of cytotoxic T lymphocytes represent an important objective for both clinical and experimental settings. Methods Here we show a simple and reproducible assay for the evaluation of antigen-specific CD8+ cytotoxic T lymphocytes based on a LysiSpot technique for the simultaneous determination of antigen-specific IFN-γ production and assessment of tumor cytolysis. The assay was developed within an experimental model of colorectal carcinoma, induced by the colorectal tumor cell line DHD-K12 that induces tumors in BDIX rats and, in turn, elicits a tumor- specific immune response. Results Using DHD-K12 cells transfected to express Escherichia coli β-galactosidase as target cells, and by the fine setting of spot colours detection, we have developed an in vitro assay that allows the recognition of cytotoxic T lymphocytes induced in BDIX rats as well as the assessment of anti-tumour cytotoxicity. The method highlighted that in the present experimental model the tumour antigen-specific immune response was bound to killing target cells in the proportion of 55%, while 45% of activated cells were not cytotoxic but released IFN-γ. Moreover in this model by an ELISPOT assay we demonstrated the specific recognition of a nonapeptide epitope called CSH-275 constitutionally express in DHD-K12 cells. Conclusions The assay proved to be highly sensitive and specific, detecting even low frequencies of cytotoxic/activated cells and providing the evaluation of cytokine-expressing T cells as well as the extent of cytotoxicity against the target cells as independent functions. This assay may represent an important tool to be adopted in experimental settings including the development of vaccines or immune therapeutic strategies
Collapse
Affiliation(s)
- Valentina Bordignon
- Laboratory of Clinical Pathology and Microbiology, San Gallicano Dermatologic Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|