1
|
Lu R, Ang YS, Cheung K, Quek KY, Sin W, Lee E, Lim SL, Yung LL, Birnbaum ME, Han J, Cheow LF, Zeming KK. iSECRETE: Integrating Microfluidics and DNA Proximity Amplification for Synchronous Single-Cell Activation and IFN-γ Secretion Profiling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309920. [PMID: 39175207 PMCID: PMC11516109 DOI: 10.1002/advs.202309920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/27/2024] [Indexed: 08/24/2024]
Abstract
Cytokines, crucial in immune modulation, impact disease progression when their secretion is dysregulated. Existing methods for profiling cytokine secretion suffer from time-consuming and labor-intensive processes and often fail to capture the dynamic nature of immune responses. Here, iSECRETE, an integrated platform that enables synchronous cell activation, wash-free, and target-responsive protein detection for single-cell IFN-γ cytokine secretion analysis within 30 min at room temperature is presented. By incorporating a DNA proximity assay (DPA) into a multifunctional microfluidic system, one-pot homogenous cytokine signal amplification, with a limit of detection of ≈50 secreted molecules per cell is achieved. iSECRETE can robustly handle various sample types that are shown. Two distinct immune activation assay modalities are demonstrated on iSECRETE. Finally, the detection of single-cell IFN-γ secretion as an activation hallmark of chimeric antigen receptor T cells within 6 h of exposure to cancer targets is shown. iSECRETE represents the fastest single-cell sample-to-result cytokine secretion assay to date, providing a powerful tool for advancing the understanding of biological phenotypes, functions, and pathways under in vivo-like conditions.
Collapse
Affiliation(s)
- Ri Lu
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
- Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingapore119077Singapore
| | - Yan Shan Ang
- Department of Chemical and Biomolecular EngineeringNational University of SingaporeSingapore117585Singapore
| | - Ka‐Wai Cheung
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
| | - Kai Yun Quek
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
| | - Wei‐Xiang Sin
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
| | - Elizabeth Lee
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
| | - Shir Lynn Lim
- National University Health SystemNational University HospitalSingapore119228Singapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Lin‐Yue Lanry Yung
- Department of Chemical and Biomolecular EngineeringNational University of SingaporeSingapore117585Singapore
| | - Michael E. Birnbaum
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Electrical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Lih Feng Cheow
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
- Department of Biomedical EngineeringNational University of SingaporeSingapore117583Singapore
| | - Kerwin Kwek Zeming
- Critical Analytics for Manufacturing of Personalised Medicine IRGSingapore‐MIT Alliance for Research and TechnologySingapore138602Singapore
| |
Collapse
|
2
|
Manohar SM. Shedding Light on Intracellular Proteins using Flow Cytometry. Cell Biochem Biophys 2024; 82:1693-1707. [PMID: 38831173 DOI: 10.1007/s12013-024-01338-1] [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] [Accepted: 05/24/2024] [Indexed: 06/05/2024]
Abstract
Intracellular protein abundance is routinely measured in mammalian cells using population-based techniques such as western blotting which fail to capture single cell protein levels or using fluorescence microscopy which is although suitable for single cell protein detection but not for rapid analysis of large no. of cells. Flow cytometry offers rapid, high-throughput, multiparameter-based analysis of intracellular protein expression in statistically significant no. of cells at single cell resolution. In past few decades, customized assays have been developed for flow cytometric detection of specific intracellular proteins. This review discusses the scope of flow cytometry for intracellular protein detection in mammalian cells along with specific applications. Technological advancements to overcome the limitations of traditional flow cytometry for the same are also discussed.
Collapse
Affiliation(s)
- Sonal M Manohar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be) University, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
3
|
Ivison S, Boucher G, Zheng G, Garcia RV, Kohen R, Bitton A, Rioux JD, Levings MK. Improving Reliability of Immunological Assays by Defining Minimal Criteria for Cell Fitness. Immunohorizons 2024; 8:622-634. [PMID: 39248805 PMCID: PMC11447670 DOI: 10.4049/immunohorizons.2300095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Human PBMC-based assays are often used as biomarkers for the diagnosis and prognosis of disease, as well as for the prediction and tracking of response to biological therapeutics. However, the development and use of PBMC-based biomarker assays is often limited by poor reproducibility. Complex immunological assays can be further complicated by variation in cell handling before analysis, especially when using cryopreserved cells. Variation in postthaw viability is further increased if PBMC isolation and cryopreservation are done more than a few hours after collection. There is currently a lack of evidence-based standards for the minimal PBMC viability or "fitness" required to ensure the integrity and reproducibility of immune cell-based assays. In this study, we use an "induced fail" approach to examine the effect of thawed human PBMC fitness on four flow cytometry-based assays. We found that cell permeability-based viability stains at the time of thawing did not accurately quantify cell fitness, whereas a combined measurement of metabolic activity and early apoptosis markers did. Investigation of the impact of different types and levels of damage on PBMC-based assays revealed that only when cells were >60-70% live and apoptosis negative did biomarker values cease to be determined by cell fitness rather than the inherent biology of the cells. These data show that, to reproducibly measure immunological biomarkers using cryopreserved PBMCs, minimal acceptable standards for cell fitness should be incorporated into the assay protocol.
Collapse
Affiliation(s)
- Sabine Ivison
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | | | - Grace Zheng
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Rosa V Garcia
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Rita Kohen
- McGill University Health Centre, Montreal, Quebec, Canada
| | - Alain Bitton
- McGill University Health Centre, Montreal, Quebec, Canada
| | - John D Rioux
- Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
do Prado Servian C, Masson LC, Fonseca SG. Phenotypic and Functional Characterization of Memory CD4 + and CD8 + T Cells After Antigenic Stimulation. Methods Mol Biol 2024; 2782:175-188. [PMID: 38622402 DOI: 10.1007/978-1-0716-3754-8_14] [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: 04/17/2024]
Abstract
The encounter of T cells with the antigen through the interaction of T cell receptors with peptides and major histocompatibility complex (MHC) molecules on the surface of antigen-presenting cells (APCs) can generate effector response and memory T cells. Memory T cells developed following infections or vaccination may persist, leading to the generation of a specific immune response upon reexposure to the same pathogen through rapid clonal proliferation and activation of effector functions. T cell memory subsets can be identified based on the expression of several membrane markers such as CCR7, CD27, and CD45RA. Using fluorescent antibodies against these markers and a flow cytometer, it is possible to perform immunophenotyping via the analysis of cell surface expression of proteins by different subpopulations such as the subsets of naïve, effector, and memory T cells as well as via the analysis of functional markers that further characterize each sample. Intracellular cytokine staining allows for the evaluation of intracellular proteins expressed in T cells in response to antigenic stimulation. This chapter presents the phenotypic and functional characterization of memory T cells after antigenic stimulation, detailing the procedures for identifying intracellular and surface protein markers. Herein, we review and present a reproducible standardized protocol using antibodies for specific markers and applying flow cytometry.
Collapse
Affiliation(s)
- Carolina do Prado Servian
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Letícia Carrijo Masson
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Simone Gonçalves Fonseca
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil.
| |
Collapse
|
5
|
Calderilla-Barbosa L, Flores-Sevilla JL. CYTOSPOT: Intracellular Staining and Detection of Cytokines by Flow Cytometer, an Alternative to LYMESPOT. Methods Mol Biol 2024; 2742:91-98. [PMID: 38165617 DOI: 10.1007/978-1-0716-3561-2_7] [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: 01/04/2024]
Abstract
Intracellular cytokine staining is a versatile technique used to analyze cytokine production in individual cells by flow cytometry. This methodology has the specific advantage of enabling the simultaneous assessment of multiple phenotypic, differentiation, and functional parameters pertaining to responding T cells. This methodology applied after short-term culture of cells, followed by fixation and permeabilization make this technique ideal for the assessment of T-cell immune responses induced by different challenges. Here we describe an intracellular staining method followed by flow cytometry after cell stimulation with immune-relevant antigens for Lyme disease.
Collapse
|
6
|
Park J, Champion JA. Development of Self-Assembled Protein Nanocage Spatially Functionalized with HA Stalk as a Broadly Cross-Reactive Influenza Vaccine Platform. ACS NANO 2023; 17:25045-25060. [PMID: 38084728 PMCID: PMC10753887 DOI: 10.1021/acsnano.3c07669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023]
Abstract
There remains a need for the development of a universal influenza vaccine, as current seasonal influenza vaccines exhibit limited protection against mismatched, mutated, or pandemic influenza viruses. A desirable approach to developing an effective universal influenza vaccine is the incorporation of highly conserved antigens in a multivalent scaffold that enhances their immunogenicity. Here, we develop a broadly cross-reactive influenza vaccine by functionalizing self-assembled protein nanocages (SAPNs) with multiple copies of the hemagglutinin stalk on the outer surface and matrix protein 2 ectodomain on the inner surface. SAPNs were generated by engineering short coiled coils, and the design was simulated by MD GROMACS. Due to the short sequences, off-target immune responses against empty SAPN scaffolds were not seen in immunized mice. Vaccination with the multivalent SAPNs induces high levels of broadly cross-reactive antibodies of only external antigens, demonstrating tight spatial control over the designed antigen placement. This work demonstrates the use of SAPNs as a potential influenza vaccine.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Julie A. Champion
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| |
Collapse
|
7
|
Badami GD, La Manna MP, Di Carlo P, Stanek O, Linhartova I, Caccamo N, Sebo P, Dieli F. Delivery of Mycobacterium tuberculosis epitopes by Bordetella pertussis adenylate cyclase toxoid expands HLA-E-restricted cytotoxic CD8 + T cells. Front Immunol 2023; 14:1289212. [PMID: 38106407 PMCID: PMC10722248 DOI: 10.3389/fimmu.2023.1289212] [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: 09/05/2023] [Accepted: 11/10/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction Tuberculosis (TB) remains the first cause of death from infection caused by a bacterial pathogen. Chemotherapy does not eradicate Mycobacterium tuberculosis (Mtb) from human lungs, and the pathogen causes a latent tuberculosis infection that cannot be prevented by the currently available Bacille Calmette Guerin (BCG) vaccine, which is ineffective in the prevention of pulmonary TB in adults. HLA-E-restricted CD8+ T lymphocytes are essential players in protective immune responses against Mtb. Hence, expanding this population in vivo or ex vivo may be crucial for vaccination or immunotherapy against TB. Methods The enzymatically inactive Bordetella pertussis adenylate cyclase (CyaA) toxoid is an effective tool for delivering peptide epitopes into the cytosol of antigen-presenting cells (APC) for presentation and stimulation of specific CD8+ T-cell responses. In this study, we have investigated the capacity of the CyaA toxoid to deliver Mtb epitopes known to bind HLA-E for the expansion of human CD8+ T cells in vitro. Results Our results show that the CyaA-toxoid containing five HLA-E-restricted Mtb epitopes causes significant expansion of HLA-E-restricted antigen-specific CD8+ T cells, which produce IFN-γ and exert significant cytotoxic activity towards peptide-pulsed macrophages. Discussion HLA-E represents a promising platform for the development of new vaccines; our study indicates that the CyaA construct represents a suitable delivery system of the HLA-E-binding Mtb epitopes for ex vivo and in vitro expansion of HLA-E-restricted CD8+ T cells inducing a predominant Tc1 cytokine profile with a significant increase of IFN-γ production, for prophylactic and immunotherapeutic applications against Mtb.
Collapse
Affiliation(s)
- Giusto D. Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, Palermo, Italy
| | - Marco P. La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, Palermo, Italy
| | - Paola Di Carlo
- Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Ondrej Stanek
- Laboratory of Molecular Biology of Bacterial Pathogen, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Irena Linhartova
- Laboratory of Molecular Biology of Bacterial Pathogen, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, Palermo, Italy
| | - Peter Sebo
- Laboratory of Molecular Biology of Bacterial Pathogen, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), AOUP Paolo Giaccone, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis (BIND), University of Palermo, Palermo, Italy
| |
Collapse
|
8
|
Erez N, Achdout H, Yahalom-Ronen Y, Adutler-Lieber S, Bar-On L, Bar-Haim E, Politi B, Vitner EB, Tamir H, Melamed S, Paran N, Israely T. Identification of T-Cell Epitopes Using a Combined In-Silico and Experimental Approach in a Mouse Model for SARS-CoV-2. Curr Issues Mol Biol 2023; 45:7944-7955. [PMID: 37886945 PMCID: PMC10605721 DOI: 10.3390/cimb45100502] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Following viral infection, T-cells are crucial for an effective immune response to intracellular pathogens, including respiratory viruses. During the COVID-19 pandemic, diverse assays were required in pre-clinical trials to evaluate the immune response following vaccination against SARS-CoV-2 and assess the response following exposure to the virus. To assess the nature and potency of the cellular response to infection or vaccination, a reliable and specific activity assay was needed. A cellular activity assay based on the presentation of short peptides (epitopes) allows the identification of T cell epitopes displayed on different alleles of the MHC, shedding light on the strength of the immune response towards antigens and aiding in antigen design for vaccination. In this report, we describe two approaches for scanning T cell epitopes on the surface glycoprotein of the SARS-CoV-2 (spike), which is utilized for attachment and entry and serves as an antigen in many vaccine candidates. We demonstrate that epitope scanning is feasible using peptide libraries or computational scanning combined with a cellular activity assay. Our scans identified four CD8 T cell epitopes, including one novel undescribed epitope. These epitopes enabled us to establish a reliable T-cell response assay, which was examined and used in various experimental mouse models for SARS-CoV-2 infection and vaccination. These approaches could potentially aid in future antigen design for vaccination and establish cellular activity assays against uncharacterized antigens of emerging pathogens.
Collapse
Affiliation(s)
- Noam Erez
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Hagit Achdout
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Yfat Yahalom-Ronen
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Shimrit Adutler-Lieber
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Liat Bar-On
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (L.B.-O.); (E.B.-H.)
| | - Erez Bar-Haim
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (L.B.-O.); (E.B.-H.)
| | - Boaz Politi
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Einat B. Vitner
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Hadas Tamir
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| | - Tomer Israely
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (H.A.); (Y.Y.-R.); (S.A.-L.); (B.P.); (E.B.V.); (H.T.); (S.M.); (N.P.)
| |
Collapse
|
9
|
De Biasi S, Paolini A, Lo Tartaro D, Gibellini L, Cossarizza A. Analysis of Antigen-Specific T and B Cells for Monitoring Immune Protection Against SARS-CoV-2. Curr Protoc 2023; 3:e636. [PMID: 36598346 DOI: 10.1002/cpz1.636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Immunological memory is the basis of protection against most pathogens. Long-living memory T and B cells able to respond to specific stimuli, as well as persistent antibodies in plasma and in other body fluids, are crucial for determining the efficacy of vaccination and for protecting from a second infection by a previously encountered pathogen. Antigen-specific cells are represented at a very low frequency in the blood, and indeed, they can be considered "rare events" present in the memory T-cell pool. Therefore, such events should be analyzed with careful attention. In the last 20 years, different methods, mostly based upon flow cytometry, have been developed to identify such rare antigen-specific cells, and the COVID-19 pandemic has given a dramatic impetus to characterize the immune response against the virus. In this regard, we know that the identification, enumeration, and characterization of SARS-CoV-2-specific T and B cells following infection and/or vaccination require i) the use of specific peptides and adequate co-stimuli, ii) the use of appropriate inhibitors to avoid nonspecific activation, iii) the setting of appropriate timing for stimulation, and iv) the choice of adequate markers and reagents to identify antigen-specific cells. Optimization of these procedures allows not only determination of the magnitude of SARS-CoV-2-specific responses but also a comparison of the effects of different combinations of vaccines or determination of the response provided by so-called "hybrid immunity," resulting from a combination of natural immunity and vaccine-generated immunity. Here, we present two methods that are largely used to monitor the response magnitude and phenotype of SARS-CoV-2-specific T and B cells by polychromatic flow cytometry, along with some tips that can be useful for the quantification of these rare events. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Identification of antigen-specific T cells Basic Protocol 2: Identification of antigen-specific B cells.
Collapse
Affiliation(s)
- Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, via Campi, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, via Campi, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, via Campi, Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, via Campi, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, via Campi, Modena, Italy.,Istituto Nazionale per le Ricerche Cardiovascolari - INRC, via Irnerio, Bologna, Italy
| |
Collapse
|
10
|
Corbière V, Lambert EE, Rodesch M, van Gaans-van den Brink JAM, Misiak A, Simonetti E, Van Praet A, Godefroid A, Diavatopoulos DA, van Els CACM, Mascart F. A semi high-throughput whole blood-based flow cytometry assay to detect and monitor Bordetella pertussis-specific Th1, Th2 and Th17 responses. Front Immunol 2023; 14:1101366. [PMID: 36814927 PMCID: PMC9939445 DOI: 10.3389/fimmu.2023.1101366] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/24/2023] [Indexed: 02/09/2023] Open
Abstract
Introduction The characterization of B. pertussis (Bp) antigen-specific CD4+ T cell cytokine responses should be included in the evaluation of immunogenicity of pertussis vaccines but is often hindered by the lack of standardized robust assays. Methods To overcome this limitation, we developed a two-step assay comprising a short-term stimulation of fresh whole blood with Bp antigens and cryopreservation of the stimulated cells, followed later on by batch-wise intracellular cytokine analysis by flow cytometry. Blood samples collected from recently acellular (aP) vaccine boosted subjects with a whole-cell- or aP-primed background was incubated for 24 hrs with Pertussis toxin, Filamentous hemagglutinin or a Bp lysate (400µl per stimulation). Antigen-specific IFN-γ-, IL-4/IL-5/IL-13-, IL-17A/IL-17F- and/or IL-22-producing CD4+ T cells were quantified by flow cytometry to reveal Th1, Th2, and Th17-type responses, respectively. The frequencies of IFN-γ-producing CD8+ T cells were also analyzed. Results We demonstrate high reproducibility of the Bp-specific whole blood intracellular staining assay. The results obtained after cryopreservation of the stimulated and fixed cells were very well correlated to those obtained without cryopreservation, an approach used in our previously published assay. Optimization resulted in high sensitivity thanks to very low non-specific backgrounds, with reliable detection of Bp antigen-specific Th1, Th2 and Th17-type CD4+ T cells, in the lowest range frequency of 0.01-0.03%. Bp antigen-specific IFN-γ+ CD8+ T lymphocytes were also detected. This test is easy to perform, analyse and interpret with the establishment of strict criteria defining Bp antigen responses. Discussion Thus, this assay appears as a promising test for evaluation of Bp antigen-specific CD4+ T cells induced by current and next generation pertussis vaccines.
Collapse
Affiliation(s)
- Véronique Corbière
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Eleonora E Lambert
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Marine Rodesch
- Department of Paediatrics, Cliniques Universitaires de Bruxelles, Hôpital Erasme, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | | | - Alicja Misiak
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Elles Simonetti
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anne Van Praet
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Audrey Godefroid
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Dimitri A Diavatopoulos
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cécile A C M van Els
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands.,Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Françoise Mascart
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | | |
Collapse
|
11
|
Railean V, Buszewski B. Flow Cytometry - Sophisticated Tool for Basic Research or/and Routine Diagnosis; Impact of the Complementarity in Both Pre- as Well as Clinical Studies. Crit Rev Anal Chem 2022; 54:2087-2109. [PMID: 36576036 DOI: 10.1080/10408347.2022.2154596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Flow cytometry is a sophisticated technology used widely in both basic research and as a routine tool in clinical diagnosis. The technology has progressed from single parameter detection in the 1970s and 1980s to high end multicolor analysis, with currently 30 parameters detected simultaneously, allowing the identification and purification of rare subpopulations of cells of interest. Flow cytometry continues to evolve and expand to facilitate the investigation of new diagnostic and therapeutic avenues. The present review gives an overview of basic theory and instrumentation, presents and compares the advantages and disadvantages of conventional, spectral and imaging flow cytometry as well as mass cytometry. Current methodologies and applications in both research, pre- and clinical settings are discussed, as well as potential limitations and future evolution. This finding encourages the reader to promote such relationship between basic science, diagnosis and multidisciplinary approach since the standard methods have limitations (e.g., in differentiating the cells after staining). Moreover, such path inspires future cytometry specialists develop new/alternative frontiers between pre- and clinical diagnosis and be more flexible in designing the study for both human as well as veterinary medicine.
Collapse
Affiliation(s)
- Viorica Railean
- Department of Infectious, Invasive Diseases and Veterinary Administration, Institute of Veterinary Medicine, Toruń, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Torun, Poland
| | - Bogusław Buszewski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Torun, Poland
- Department of Environmental Chemistry and Bioanalysis, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
12
|
Bhuiyan TR, Al Banna H, Kaisar MH, Karmakar PC, Hakim A, Akter A, Ahmed T, Tauheed I, Islam S, Hasnat MA, Sumon MA, Rashed A, Ghosh S, Clemens JD, Banu S, Shirin T, Weiskopf D, Sette A, Chowdhury F, Qadri F. Correlation of antigen-specific immune response with disease severity among COVID-19 patients in Bangladesh. Front Immunol 2022; 13:929849. [PMID: 36248882 PMCID: PMC9554593 DOI: 10.3389/fimmu.2022.929849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a protean disease causing different degrees of clinical severity including fatality. In addition to humoral immunity, antigen-specific T cells may play a critical role in defining the protective immune response against SARS-CoV-2, the virus that causes this disease. As a part of a longitudinal cohort study in Bangladesh to investigate B and T cell-specific immune responses, we sought to evaluate the activation-induced marker (AIM) and the status of different immune cell subsets during a COVID-19 infection. We analyzed a total of 115 participants, which included participants with asymptomatic, mild, moderate, and severe clinical symptoms. We observed decreased mucosal-associated invariant T (MAIT) cell frequency on the initial days of the COVID-19 infection in symptomatic patients compared to asymptomatic patients. However, natural killer (NK) cells were found to be elevated in symptomatic patients just after the onset of the disease compared to both asymptomatic patients and healthy individuals. Moreover, we found a significant increase of AIM+ (both OX40+CD137+ and OX40+CD40L+) CD4+ T cells in moderate and severe COVID-19 patients in response to SARS-CoV-2 peptides (especially spike peptides) compared to pre-pandemic controls who are unexposed to SARS-CoV-2. Notably, we did not observe any significant difference in the CD8+ AIMs (CD137+CD69+), which indicates the exhaustion of CD8+ T cells during a COVID-19 infection. These findings suggest that patients who recovered from moderate and severe COVID-19 were able to mount a strong CD4+ T-cell response against shared viral determinants that ultimately induced T cells to mount further immune responses to SARS-CoV-2.
Collapse
Affiliation(s)
- Taufiqur Rahman Bhuiyan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Hasan Al Banna
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - M. Hasanul Kaisar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Polash Chandra Karmakar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Al Hakim
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
- Department of Genetic Engineering and Biotechnology, Jagannath University, Dhaka, Bangladesh
| | - Afroza Akter
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Tasnuva Ahmed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Imam Tauheed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Shaumik Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Mohammad Abul Hasnat
- Department of Cardiology, Department of Oncology, Kurmitola General Hospital, Dhaka, Bangladesh
| | - Mostafa Aziz Sumon
- Department of Cardiology, Department of Oncology, Kurmitola General Hospital, Dhaka, Bangladesh
| | - Asif Rashed
- Department of Microbiology, Department of Medicine, Mugda Medical College and Hospital, Dhaka, Bangladesh
| | - Shuvro Ghosh
- Department of Microbiology, Department of Medicine, Mugda Medical College and Hospital, Dhaka, Bangladesh
| | - John D. Clemens
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
- Department of Epidemiology, University of California Los Angeles (UCLA) Fielding School of Public Health, Los Angeles, CA, United States
- International Vaccine Institute, Seoul, South Korea
| | - Sayera Banu
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Tahmina Shirin
- Institute of Epidemiology, Disease Control and Research (IEDCR), Dhaka, Bangladesh
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Fahima Chowdhury
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
- *Correspondence: Firdausi Qadri,
| |
Collapse
|
13
|
Halliday A, Masonou T, Tolosa-Wright MR, Guo Y, Hoang L, Parker R, Boakye A, Takwoingi Y, Badhan A, Jain P, Marwah I, Berrocal-Almanza LC, Deeks J, Beverley P, Kon OM, Lalvani A. Defining the Role of Cellular Immune Signatures in Diagnostic Evaluation of Suspected Tuberculosis. J Infect Dis 2022; 225:1632-1641. [PMID: 34331451 PMCID: PMC9071295 DOI: 10.1093/infdis/jiab311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Diagnosis of paucibacillary tuberculosis (TB) including extrapulmonary TB is a significant challenge, particularly in high-income, low-incidence settings. Measurement of Mycobacterium tuberculosis (Mtb)-specific cellular immune signatures by flow cytometry discriminates active TB from latent TB infection (LTBI) in case-control studies; however, their diagnostic accuracy and clinical utility in routine clinical practice is unknown. METHODS Using a nested case-control study design within a prospective multicenter cohort of patients presenting with suspected TB in England, we assessed diagnostic accuracy of signatures in 134 patients who tested interferon-gamma release assay (IGRA)-positive and had final diagnoses of TB or non-TB diseases with coincident LTBI. Cellular signatures were measured using flow cytometry. RESULTS All signatures performed less well than previously reported. Only signatures incorporating measurement of phenotypic markers on functional Mtb-specific CD4 T cells discriminated active TB from non-TB diseases with LTBI. The signatures measuring HLA-DR+IFNγ + CD4 T cells and CD45RA-CCR7-CD127- IFNγ -IL-2-TNFα + CD4 T cells performed best with 95% positive predictive value (95% confidence interval, 90-97) in the clinically challenging subpopulation of IGRA-positive but acid-fast bacillus (AFB) smear-negative TB suspects. CONCLUSIONS Two cellular immune signatures could improve and accelerate diagnosis in the challenging group of patients who are IGRA-positive, AFB smear-negative, and have paucibacillary TB.
Collapse
Affiliation(s)
- Alice Halliday
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Bristol Vaccine Centre, Cellular and Molecular Medicine, Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Tereza Masonou
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Mica R Tolosa-Wright
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| | - Yanping Guo
- St Mary’s Flow Cytometry Facility, Imperial College London, London, United Kingdom
| | - Long Hoang
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Robert Parker
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Aime Boakye
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
- NIHR Imperial Clinical Research Facility, Hammersmith Hospital, London, United Kingdom
| | - Yemisi Takwoingi
- Institute of Applied Health Research, University of Birmingham and NIHR Birmingham, Biomedical Research Centre (University Hospital Birmingham NHS Foundation Trust and University of Birmingham), Birmingham, United Kingdom
| | - Amarjit Badhan
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| | - Pooja Jain
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| | - Ishita Marwah
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| | - Luis C Berrocal-Almanza
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| | - Jonathan Deeks
- Institute of Applied Health Research, University of Birmingham and NIHR Birmingham, Biomedical Research Centre (University Hospital Birmingham NHS Foundation Trust and University of Birmingham), Birmingham, United Kingdom
| | - Peter Beverley
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| | - Onn Min Kon
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
- St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ajit Lalvani
- TB Research Centre, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Health Protection Research Unit in Respiratory Infections, Imperial College London, London, United Kingdom
| |
Collapse
|
14
|
CD4 T Cell-Mediated Immune Control of Cytomegalovirus Infection in Murine Salivary Glands. Pathogens 2021; 10:pathogens10121531. [PMID: 34959486 PMCID: PMC8704252 DOI: 10.3390/pathogens10121531] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/24/2022] Open
Abstract
CD4 T cells are well known for their supportive role in CD8 T cell and B cell responses during viral infection. However, during murine cytomegalovirus (MCMV) infection in the salivary glands (SGs), CD4 T cells exhibit direct antiviral effector functions to control the infection. In this mucosal organ, opposed to other infected tissues, MCMV establishes a sustained lytic replication that lasts for several weeks. While the protective function of CD4 T cells is exerted through the production of the pro-inflammatory cytokines interferon gamma (IFNγ) and tumor necrosis factor alpha (TNF), the reasons for their markedly delayed control of lytic MCMV infection remain elusive. Here, we review the current knowledge on the dynamics and mechanisms of the CD4 T cell-mediated control of MCMV-infected SGs, including their localization in the SG in relation to MCMV infected cells and other immune cells, their mode of action, and their regulation.
Collapse
|
15
|
Shekarkar Azgomi M, La Manna MP, Badami GD, Ragonese P, Trizzino A, Dieli F, Caccamo N. A Rapid and Simple Multiparameter Assay to Quantify Spike-Specific CD4 and CD8 T Cells after SARS-CoV-2 Vaccination: A Preliminary Report. Biomedicines 2021; 9:1576. [PMID: 34829805 PMCID: PMC8615821 DOI: 10.3390/biomedicines9111576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
mRNA and Adenovirus vaccines for COVID-19 are used to induce humoral and cell-mediated immunity, with the aim to generate both SARS-CoV-2 B and T memory cells. In present study, we described a simple assay to detect and quantify Spike-specific CD4+ and CD8+ T cell responses induced by vaccination in healthy donors and in subjects with B cell compart impairment, in which antibody response is absent due to primary immunodeficiencies or CD20 depleting therapy. We detect and quantified memory T cell immune responses against SARS-CoV-2 evocated by vaccination in both groups, irrespective to the humoral response. Furthermore, we identified TNF-α as the main cytokine produced by T memory cells, after antigen-specific stimulation in vitro, that could be considered, other than IFN-γ, an additional biomarker of induction of T memory cells upon vaccination. Further studies on the vaccine-induced T cell responses could be crucial, not only in healthy people but also in immunocompromised subjects, where antigen specific T cells responses play a protective role against SARS-CoV-2.
Collapse
Affiliation(s)
- Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnostic and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy; (M.S.A.); (M.P.L.M.); (G.D.B.); (F.D.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, 90127 Palermo, Italy;
| | - Marco Pio La Manna
- Central Laboratory of Advanced Diagnostic and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy; (M.S.A.); (M.P.L.M.); (G.D.B.); (F.D.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, 90127 Palermo, Italy;
| | - Giusto Davide Badami
- Central Laboratory of Advanced Diagnostic and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy; (M.S.A.); (M.P.L.M.); (G.D.B.); (F.D.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, 90127 Palermo, Italy;
| | - Paolo Ragonese
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, 90127 Palermo, Italy;
| | - Antonino Trizzino
- Department of Pediatric Hematology and Oncology, A.R.N.A.S. Civico Di Cristina and Benfratelli Hospital, 90127 Palermo, Italy;
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnostic and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy; (M.S.A.); (M.P.L.M.); (G.D.B.); (F.D.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, 90127 Palermo, Italy;
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnostic and Biomedical Research (CLADIBIOR), University of Palermo, 90127 Palermo, Italy; (M.S.A.); (M.P.L.M.); (G.D.B.); (F.D.)
- Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
16
|
Liu C, Chu D, Kalantar‐Zadeh K, George J, Young HA, Liu G. Cytokines: From Clinical Significance to Quantification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004433. [PMID: 34114369 PMCID: PMC8336501 DOI: 10.1002/advs.202004433] [Citation(s) in RCA: 269] [Impact Index Per Article: 89.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/26/2021] [Indexed: 05/24/2023]
Abstract
Cytokines are critical mediators that oversee and regulate immune and inflammatory responses via complex networks and serve as biomarkers for many diseases. Quantification of cytokines has significant value in both clinical medicine and biology as the levels provide insights into physiological and pathological processes and can be used to aid diagnosis and treatment. Cytokines and their clinical significance are introduced from the perspective of their pro- and anti-inflammatory effects. Factors affecting cytokines quantification in biological fluids, native levels in different body fluids, sample processing and storage conditions, sensitivity to freeze-thaw, and soluble cytokine receptors are discussed. In addition, recent advances in in vitro and in vivo assays, biosensors based on different signal outputs and intracellular to extracellular protein expression are summarized. Various quantification platforms for high-sensitivity and reliable measurement of cytokines in different scenarios are discussed, and commercially available cytokine assays are compared. A discussion of challenges in the development and advancement of technologies for cytokine quantification that aim to achieve real-time multiplex cytokine analysis for point-of-care situations applicable for both biomedical research and clinical practice are discussed.
Collapse
Affiliation(s)
- Chao Liu
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | - Dewei Chu
- School of Materials Science and EngineeringUniversity of New South WalesSydneyNSW2052Australia
| | | | - Jacob George
- Storr Liver CentreWestmead Institute of Medical ResearchUniversity of Sydney and Department of Gastroenterology and HepatologyWestmead HospitalWestmeadNSW2145Australia
| | - Howard A. Young
- Laboratory of Cancer ImmunometabolismCenter for Cancer ResearchNational Cancer Institute at FrederickFrederickMD21702USA
| | - Guozhen Liu
- School of Life and Health SciencesThe Chinese University of Hong KongShenzhen518172P. R. China
- Graduate School of Biomedical EngineeringUniversity of New South WalesSydneyNSW2052Australia
| |
Collapse
|
17
|
Lu M, Panebra A, Kim WH, Lillehoj HS. Characterization of immunological properties of chicken chemokine CC motif ligand 5 using new monoclonal antibodies. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104023. [PMID: 33497732 DOI: 10.1016/j.dci.2021.104023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
CCL5 (formerly RANTES) belongs to the CC (or β) chemokine family and is associated with a plethora of inflammatory disorders and pathologic states. CCL5 is mainly produced and secreted by T cells, macrophages, epithelial cells, and fibroblasts and acts as a chemoattractant to recruit effector cells to the inflammation sites. Chicken CCL5 (chCCL5) protein is closely related to avian CCL5 orthologs but distinct from mammalian orthologs, and its modulatory roles in the immune response are largely unknown. The present work was undertaken to characterize the immunological properties of chCCL5 using the new sets of anti-chCCL5 mouse monoclonal antibodies (mAbs). Eight different mAbs (6E11, 6H1, 8H11, 11G1, 11G11, 12H1, 13D1, and 13G3) were characterized for their specificity and binding ability toward chCCL5. Two (13G3 and 6E11) of them were selected to detect native chCCL5 in chCCL5-specific antigen-capture ELISA. Using 13G3 and 6E11 as capture and detection antibodies, respectively, the ELISA system detected serum chCCL5 secretions in Clostridium perfringens- and Eimeria-infected chickens. The intracellular expressions of chCCL5 in primary cells or cell lines derived from chickens were validated in immunocytochemistry and flow cytometry assays using both 13G3 and 6E11 mAbs. Furthermore, 6E11, but not 13G3, neutralized chCCL5-induced chemotaxis in vitro using chicken PBMCs. These molecular characteristics of chCCL5 demonstrate the potential application of anti-chCCL5 mAbs and CCL5-specific antigen-capture detection ELISA for detecting native chCCL5 in biological samples. The availability of these new immunological tools will be valuable for fundamental and applied studies in avian species.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA
| | - Alfredo Panebra
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA
| | - Woo H Kim
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture, Beltsville, MD, 20705, USA.
| |
Collapse
|
18
|
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
|
19
|
Moris P, Bellanger A, Ofori-Anyinam O, Jongert E, Yarzabal Rodriguez JP, Janssens M. Whole blood can be used as an alternative to isolated peripheral blood mononuclear cells to measure in vitro specific T-cell responses in human samples. J Immunol Methods 2021; 492:112940. [PMID: 33493551 DOI: 10.1016/j.jim.2020.112940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/30/2020] [Accepted: 12/07/2020] [Indexed: 11/19/2022]
Abstract
Vaccinology is confronted with diseases for which the control of T-cell responses by the vaccine is essential. Among the assays that have been designed to assess T-cell responses, intracellular cytokine staining (ICS) combined with flow cytometry is well-suited in the frame of clinical trials. This assay can be used starting from isolated peripheral blood mononuclear cells (PBMC) or from whole blood (WB), but firm equivalence between the two sample preparation methods has yet to be established. Therefore, we compared both methods by analyzing the frequency of antigen-specific CD4+ T cells expressing at least two of four immune markers in human samples taken from two independent clinical trials (NCT00397943 and NCT00805389) with a qualified ICS assay. In the first study, M72-specific CD4+ T-cell responses were analyzed using WB-ICS and PBMC-ICS in 293 samples. Of these, 128 were double positive (value ≥ lower limit of quantification [LLOQ] with both methods), 130 were double negative and only 35 sample results were discordant, leading to an overall agreement of 88.05%. When analyzing the 128 double positive samples, it was found that the geometric mean of ratios (GMR) for paired observations was 0.98, which indicates a very good alignment between the two methods. The Deming regression fitted between the methods also showed a good correlation with an estimated slope being 1.1085. In the second study, HBsAg-specific CD4+ T-cell responses were analyzed in 371 samples. Of these, 100 were double positive, 195 were double negative and 76 sample results were discordant, leading to an overall agreement of 79.51%. The GMR for paired observations was equal to 1.20, caused by a trend for overestimation in favor of the WB samples in the very high frequencies. The estimated slope of the Deming regression was 1.3057. In conclusion, we demonstrated that WB and PBMC methods of sample collection led to statistically concordant ICS results, indicating that WB-ICS is a suitable alternative to PBMC-ICS to analyze clinical trial samples.
Collapse
Affiliation(s)
| | | | | | - Erik Jongert
- GSK, Rue de l'Institut 89, 1330 Rixensart, Belgium.
| | | | | |
Collapse
|
20
|
Choi JR, Lee JH, Xu A, Matthews K, Xie S, Duffy SP, Ma H. Monolithic hydrogel nanowells-in-microwells enabling simultaneous single cell secretion and phenotype analysis. LAB ON A CHIP 2020; 20:4539-4551. [PMID: 33201962 DOI: 10.1039/d0lc00965b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cytokine secretion is a form of cellular communication that regulates a wide range of biological processes. A common approach for measuring cytokine secretion from single cells is to confine individual cells in arrays of nanoliter wells (nanowells) fabricated using polydimethylsiloxane. However, this approach cannot be easily integrated in standard microwell plates in order to take advantage of high-throughput infrastructure for automated and multiplexed analysis. Here, we used laser micropatterning to fabricate monolithic hydrogel nanowells inside wells in a microwell plate (microwells) using polyethylene glycol diacrylate (PEGDA). This approach produces high-aspect ratio nanowells that retain cells and beads during reagent exchange, enabling simultaneous profiling of single cell secretion and phenotyping via immunostaining. To limit contamination between nanowells, we used methylcellulose as a media additive to reduce diffusion distance. Patterning nanowells monolithically in microwells also dramatically increases density, providing ∼1200 nanowells per microwell in a microwell plate. Using this approach, we profiled IL-8 secretion from single MDA-MB-231 cells, which showed significant heterogeneity. We further profiled the polarization of THP-1 cells into M1 and M2 macrophages, along with their associated IL-1β and CCL-22 secretion profiles. These results demonstrate the potential to use this approach for high-throughput secretion and phenotype analysis on single cells.
Collapse
Affiliation(s)
- Jane Ru Choi
- Department of Mechanical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada
| | - Jeong Hyun Lee
- Department of Mechanical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada
| | - Alec Xu
- Department of Mechanical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada
| | - Kerryn Matthews
- Department of Mechanical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada
| | - Shuyong Xie
- Department of Mechanical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada
| | - Simon P Duffy
- Department of Mechanical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada and British Columbia Institute of Technology, Canada
| | - Hongshen Ma
- Department of Mechanical Engineering, University of British Columbia, Canada. and Centre for Blood Research, University of British Columbia, Canada and School of Biomedical Engineering, University of British Columbia, Canada and Vancouver Prostate Centre, Vancouver General Hospital, Canada
| |
Collapse
|
21
|
Rozot V, Nemes E, Geldenhuys H, Musvosvi M, Toefy A, Rantangee F, Makhethe L, Erasmus M, Bilek N, Mabwe S, Finak G, Fulp W, Ginsberg AM, Hokey DA, Shey M, Gurunathan S, DiazGranados C, Bekker LG, Hatherill M, Scriba TJ. Multidimensional analyses reveal modulation of adaptive and innate immune subsets by tuberculosis vaccines. Commun Biol 2020; 3:563. [PMID: 33037320 PMCID: PMC7547090 DOI: 10.1038/s42003-020-01288-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
We characterize the breadth, function and phenotype of innate and adaptive cellular responses in a prevention of Mycobacterium tuberculosis infection trial. Responses are measured by whole blood intracellular cytokine staining at baseline and 70 days after vaccination with H4:IC31 (subunit vaccine containing Ag85B and TB10.4), Bacille Calmette-Guerin (BCG, a live attenuated vaccine) or placebo (n = ~30 per group). H4:IC31 vaccination induces Ag85B and TB10.4-specific CD4 T cells, and an unexpected NKTlike subset, that expresses IFN-γ, TNF and/or IL-2. BCG revaccination increases frequencies of CD4 T cell subsets that either express Th1 cytokines or IL-22, and modestly increases IFNγ-producing NK cells. In vitro BCG re-stimulation also triggers responses by donor-unrestricted T cells, which may contribute to host responses against mycobacteria. BCG, which demonstrated efficacy against sustained Mycobacterium tuberculosis infection, modulates multiple immune cell subsets, in particular conventional Th1 and Th22 cells, which should be investigated in discovery studies of correlates of protection.
Collapse
Affiliation(s)
- Virginie Rozot
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Hennie Geldenhuys
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Asma Toefy
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Frances Rantangee
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Lebohang Makhethe
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mzwandile Erasmus
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Simbarashe Mabwe
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Greg Finak
- Fred Hutchinson Cancer Research Center (FHCRC), Seattle, WA, USA
| | - William Fulp
- Fred Hutchinson Cancer Research Center (FHCRC), Seattle, WA, USA
| | | | | | - Muki Shey
- Aeras South Africa Endpoint Assay Laboratory, Cape Town, South Africa
| | | | | | - Linda-Gail Bekker
- The Desmond Tutu HIV Centre, University of Cape Town, Cape Town, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
22
|
Sudbury EL, Clifford V, Messina NL, Song R, Curtis N. Mycobacterium tuberculosis-specific cytokine biomarkers to differentiate active TB and LTBI: A systematic review. J Infect 2020; 81:873-881. [PMID: 33007340 DOI: 10.1016/j.jinf.2020.09.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/21/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES New tests are needed to overcome the limitations of existing immunodiagnostic tests for tuberculosis (TB) infection, including their inability to differentiate between active TB and latent TB infection (LTBI). This review aimed to identify the most promising cytokine biomarkers for use as stage-specific markers of TB infection. METHODS A systematic review was done using electronic databases to identify studies that have investigated Mycobacterium tuberculosis (MTB)-specific cytokine responses as diagnostic tools to differentiate between LTBI and active TB. RESULTS The 56 studies included in this systematic review measured the MTB-specific responses of 100 cytokines, the most frequently studied of which were IFN-γ, IL-2, TNF-α, IP-10, IL-10 and IL-13. Ten studies assessed combinations of cytokines, most commonly IL-2 and IFN-γ. For most cytokines, findings were heterogenous between studies. The variation in results likely relates to differences in the study design and laboratory methods, as well as participant and environmental factors. CONCLUSIONS Although several cytokines show promise as stage-specific markers of TB infection, this review highlights the need for further well-designed studies, in both adult and paediatric populations, to establish which cytokine(s) will be of most use in a new generation of immunodiagnostic tests.
Collapse
Affiliation(s)
- Eva L Sudbury
- Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia.
| | - Vanessa Clifford
- Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital, Parkville, Australia.
| | - Nicole L Messina
- Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia.
| | - Rinn Song
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the National Institute for Health Research Oxford Biomedical Research Centre, Oxford, UK; Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital, Parkville, Australia.
| |
Collapse
|
23
|
Abstract
The immune system is composed of heterogeneous populations of immune cells that regulate physiological processes and protect organisms against diseases. Single cell technologies have been used to assess immune cell responses at the single cell level, which are crucial for identifying the causes of diseases and elucidating underlying biological mechanisms to facilitate medical therapy. In the present review we first discuss the most recent advances in the development of single cell technologies to investigate cell signaling, cell-cell interactions and cell migration. Each technology's advantages and limitations and its applications in immunology are subsequently reviewed. The latest progress toward commercialization, the remaining challenges and future perspectives for single cell technologies in immunology are also briefly discussed.
Collapse
Affiliation(s)
- Jane Ru Choi
- Centre for Blood Research, Life Sciences Centre, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Mechanical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
24
|
Lu M, Kim WH, Lillehoj HS, Li C. Development and characterization of novel mouse monoclonal antibodies against chicken chemokine CC motif ligand 4. Vet Immunol Immunopathol 2020; 227:110091. [PMID: 32682170 DOI: 10.1016/j.vetimm.2020.110091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/28/2020] [Accepted: 07/07/2020] [Indexed: 11/19/2022]
Abstract
Chemokine (C-C motif) ligand (CCL) 4 is a CC chemokine subfamily member defined by the sequential positioning of conserved cysteine residues. Upon the binding of G-protein-coupled receptors on the cell surface, CCL4 mediates a diverse set of biological processes including chemotaxis, tumorigenesis, homeostasis and thymopoiesis. Although the physiological roles of mammalian CCL4s were elucidated >20 years ago, there is limited information on the biological activities of chicken CCL4 (chCCL4). In the present study, we developed and characterized mouse monoclonal antibodies (mAbs) against chCCL4 to characterize better the immunological properties of chCCL4. Out of initial screening of >400 clones, two mAbs detecting chCCL4, 1A12 and 15D9, were identified and characterized using western blotting and chCCL4-specific antigen-capture enzyme-linked immunosorbent assay, and their neutralizing activity was validated by chCCL4-induced peripheral blood mononuclear cell chemotaxis assay. Furthermore, the intracellular expression of chCCL4 in various chicken cells by immunocytochemistry and flow cytometry was confirmed using 1A12 and 15D9 mAbs. These results collectively indicate that 1A12 and 15D9 mAbs specifically detect chicken CCL4 and they will be valuable immune reagents for basic and applied studies in avian immunology.
Collapse
Affiliation(s)
- Mingmin Lu
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| | - Woo H Kim
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| | - Charles Li
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Beltsville, MD, 20705, USA.
| |
Collapse
|
25
|
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
|
26
|
Estévez O, Anibarro L, Garet E, Martínez A, Pena A, Barcia L, Peleteiro M, González-Fernández Á. Multi-parameter flow cytometry immunophenotyping distinguishes different stages of tuberculosis infection. J Infect 2020; 81:57-71. [PMID: 32330526 DOI: 10.1016/j.jinf.2020.03.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To identify new potential host biomarkers in blood to discriminate between active TB patients, uninfected (NoTBI) and latently infected contacts (LTBI). METHODS A blood cell count was performed to study parent leukocyte populations. Peripheral blood mononuclear cells (PBMCs) were isolated and a multi-parameter flow cytometry assay was conducted to study the distribution of basal and Mycobacterium tuberculosis (Mtb)-stimulated lymphocytes. Differences between groups and the area under the ROC curve (AUC) were investigated to assess the diagnostic accuracy. RESULTS Active TB patients presented higher Monocyte-to-lymphocyte and Neutrophil-to-lymphocyte ratios than LTBI and NoTBI contacts (p<0.0001; AUC>0.8). Lymphocyte subsets with differences (p >0.05; AUC >0.7) between active TB and both contact groups include the basal distribution of Th1/Th2 ratio, Th1-Th17, CD4+ Central Memory (TCM) or MAIT cells. Expression of CD154 is increased in Mtb-activated CD4+ TCM and Effector Memory T cells in active TB and LTBI compared to NoTBI. In CD4+T cells, expression of CD154 showed a higher accuracy than IFNγ to discriminate Mtb-specific activation. CONCLUSIONS We identified different cell subsets with potential use in tuberculosis diagnosis. Among them, distribution of CD4 TCM cells and their expression of CD154 after Mtb-activation are the most promising candidates.
Collapse
Affiliation(s)
- Olivia Estévez
- CINBIO, Universidade de Vigo, Immunology Group, Campus universitario Lagoas, Marcosende, 36310 Vigo, Spain; Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Spain
| | - Luis Anibarro
- Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Spain; GEIM -Grupo de Estudio de Micobacterias de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica, Spain; Tuberculosis Unit, Department of Infectious Diseases and Internal Medicine, Pontevedra Hospital Complex, Pontevedra, Spain
| | - Elina Garet
- CINBIO, Universidade de Vigo, Immunology Group, Campus universitario Lagoas, Marcosende, 36310 Vigo, Spain; Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Spain
| | - Amparo Martínez
- CINBIO, Universidade de Vigo, Immunology Group, Campus universitario Lagoas, Marcosende, 36310 Vigo, Spain; Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Spain
| | - Alberto Pena
- Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Spain; Tuberculosis Unit, Department of Infectious Diseases and Internal Medicine, Pontevedra Hospital Complex, Pontevedra, Spain
| | - Laura Barcia
- Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Spain; Tuberculosis Unit, Department of Infectious Diseases and Internal Medicine, Pontevedra Hospital Complex, Pontevedra, Spain
| | - Mercedes Peleteiro
- CINBIO, Universidade de Vigo, Flow Cytometry Core Facility, Campus universitario Lagoas, Marcosende, 36310 Vigo, Spain
| | - África González-Fernández
- CINBIO, Universidade de Vigo, Immunology Group, Campus universitario Lagoas, Marcosende, 36310 Vigo, Spain; Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), Spain.
| |
Collapse
|
27
|
Abstract
Mass cytometry (MC) is a powerful research tool enabling high-dimensional analysis of single cells in suspension and within tissue sections following laser ablation. Here we describe the procedure of titrating metal-conjugated antibodies, to ensure that optimal levels of staining are achieved while minimizing nonspecific signals that may occur at high concentrations.
Collapse
|
28
|
Botafogo V, Pérez-Andres M, Jara-Acevedo M, Bárcena P, Grigore G, Hernández-Delgado A, Damasceno D, Comans S, Blanco E, Romero A, Arriba-Méndez S, Gastaca-Abasolo I, Pedreira CE, van Gaans-van den Brink JAM, Corbiere V, Mascart F, van Els CACM, Barkoff AM, Mayado A, van Dongen JJM, Almeida J, Orfao A. Age Distribution of Multiple Functionally Relevant Subsets of CD4+ T Cells in Human Blood Using a Standardized and Validated 14-Color EuroFlow Immune Monitoring Tube. Front Immunol 2020; 11:166. [PMID: 32174910 PMCID: PMC7056740 DOI: 10.3389/fimmu.2020.00166] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
CD4+ T cells comprise multiple functionally distinct cell populations that play a key role in immunity. Despite blood monitoring of CD4+ T-cell subsets is of potential clinical utility, no standardized and validated approaches have been proposed so far. The aim of this study was to design and validate a single 14-color antibody combination for sensitive and reproducible flow cytometry monitoring of CD4+ T-cell populations in human blood to establish normal age-related reference values and evaluate the presence of potentially altered profiles in three distinct disease models-monoclonal B-cell lymphocytosis (MBL), systemic mastocytosis (SM), and common variable immunodeficiency (CVID). Overall, 145 blood samples from healthy donors were used to design and validate a 14-color antibody combination based on extensive reagent testing in multiple cycles of design-testing-evaluation-redesign, combined with in vitro functional studies, gene expression profiling, and multicentric evaluation of manual vs. automated gating. Fifteen cord blood and 98 blood samples from healthy donors (aged 0-89 years) were used to establish reference values, and another 25 blood samples were evaluated for detecting potentially altered CD4 T-cell subset profiles in MBL (n = 8), SM (n = 7), and CVID (n = 10). The 14-color tube can identify ≥89 different CD4+ T-cell populations in blood, as validated with high multicenter reproducibility, particularly when software-guided automated (vs. manual expert-based) gating was used. Furthermore, age-related reference values were established, which reflect different kinetics for distinct subsets: progressive increase of naïve T cells, T-helper (Th)1, Th17, follicular helper T (TFH) cells, and regulatory T cells (Tregs) from birth until 2 years, followed by a decrease of naïve T cells, Th2, and Tregs in older children and a subsequent increase in multiple Th-cell subsets toward late adulthood. Altered and unique CD4+ T-cell subset profiles were detected in two of the three disease models evaluated (SM and CVID). In summary, the EuroFlow immune monitoring TCD4 tube allows fast, automated, and reproducible identification of ≥89 subsets of CD4+ blood T cells, with different kinetics throughout life. These results set the basis for in-depth T-cell monitoring in different disease and therapeutic conditions.
Collapse
Affiliation(s)
- Vitor Botafogo
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martín Pérez-Andres
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - María Jara-Acevedo
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Sequencing Service, NUCLEUS, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Paloma Bárcena
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Alejandro Hernández-Delgado
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
- Cytognos SL, Salamanca, Spain
| | - Daniela Damasceno
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Suzanne Comans
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Elena Blanco
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Romero
- Miguel Armijo Primary Health Care Centre, Sanidad de Castilla y León (SACYL), Salamanca, Spain
| | | | - Irene Gastaca-Abasolo
- Gynecology and Obstetrics Service, University Hospital of Salamanca, Salamanca, Spain
| | - Carlos Eduardo Pedreira
- Systems and Computing Department (PESC), COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | | - Véronique Corbiere
- Laboratory of Vaccinology and Mucosal Immunity, Université libre de Bruxelles (ULB), Brussels, Belgium
| | - Françoise Mascart
- Laboratory of Vaccinology and Mucosal Immunity, Université libre de Bruxelles (ULB), Brussels, Belgium
- Immunobiology Clinic, Hôpital Erasme, Brussels, Belgium
| | - Cécile A. C. M. van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Alex-Mikael Barkoff
- Institute of Biomedicine, Department of Microbiology, Virology and Immunology, University of Turku (UTU), Turku, Finland
| | - Andrea Mayado
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Jacques J. M. van Dongen
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Julia Almeida
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer (CIC) and Instituto de Biología Molecular y Celular del Cancer (IBMCC), CSIC-University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL) and Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC) (CB16/12/00400), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Chetaille Nézondet AL, Poubelle PE, Pelletier M. The evaluation of cytokines to help establish diagnosis and guide treatment of autoinflammatory and autoimmune diseases. J Leukoc Biol 2020; 108:647-657. [PMID: 32040246 DOI: 10.1002/jlb.5mr0120-218rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 12/17/2022] Open
Abstract
Our knowledge of the role of cytokines in pathologic conditions has increased considerably with the emergence of molecular and genetic studies, particularly in the case of autoinflammatory monogenic diseases. Many rare disorders, considered orphan until recently, are directly related to abnormal gene regulation, and the treatment with biologic agents (biologics) targeting cytokine receptors, intracellular signaling or specific cytokines improve the symptoms of an increasing number of chronic inflammatory diseases. As it is currently impossible to systematically conduct genetic studies for all patients with autoinflammatory and autoimmune diseases, the evaluation of cytokines can be seen as a simple, less time consuming, and less expensive alternative. This approach could be especially useful when the diagnosis of syndromes of diseases of unknown etiology remains problematic. The evaluation of cytokines could also help avoid the current trial-and-error approach, which has the disadvantages of exposing patients to ineffective drugs with possible unnecessary side effects and permanent organ damages. In this review, we discuss the various possibilities, as well as the limitations of evaluating the cytokine profiles of patients suffering from autoinflammatory and autoimmune diseases, with methods such as direct detection of cytokines in the plasma/serum or following ex vivo stimulation of PBMCs leading to the production of their cytokine secretome. The patients' secretome, combined with biomarkers ranging from genetic and epigenetic analyses to immunologic biomarkers, may help not only the diagnosis but also guide the choice of biologics for more efficient and rapid treatments.
Collapse
Affiliation(s)
- Anne-Laure Chetaille Nézondet
- Department of Medicine, Faculty of Medicine, Laval University, Québec, Canada.,Reproduction, Mother and Youth Health Axis, CHU de Québec-Université Laval Research Center, Québec, Canada
| | - Patrice E Poubelle
- Department of Medicine, Faculty of Medicine, Laval University, Québec, Canada.,Infectious and Immune Diseases Axis, CHU de Québec-Université Laval Research Center, Québec, Canada
| | - Martin Pelletier
- Infectious and Immune Diseases Axis, CHU de Québec-Université Laval Research Center, Québec, Canada.,Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, Canada.,ARThrite Research Center, Laval University, Québec, Canada
| |
Collapse
|
30
|
Pol JG, Bridle BW, Lichty BD. Detection of Tumor Antigen-Specific T-Cell Responses After Oncolytic Vaccination. Methods Mol Biol 2020; 2058:191-211. [PMID: 31486039 DOI: 10.1007/978-1-4939-9794-7_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oncolytic vaccines, which consist of recombinant oncolytic viruses (OV) encoding tumor-associated antigens (TAAs), have demonstrated potent antitumor efficacy in preclinical models and are currently evaluated in phase I/II clinical trials. On one hand, oncolysis of OV-infected malignant entities reinstates cancer immunosurveillance. On the other hand, overexpression of TAAs in infected cells further stimulates the adaptive arm of antitumor immunity. Particularly, the presence of tumor-specific CD8+ T lymphocytes within the tumor microenvironment, as well as in the periphery, has demonstrated prognostic value for cancer treatments. These effector CD8+ T cells can be detected through their production of the prototypical Tc1 cytokine: IFN-γ. The quantitative and qualitative assessment of this immune cell subset remains critical in the development process of efficient cancer vaccines, including oncolytic vaccines. The present chapter will describe a single-cell immunological assay, namely the intracellular cytokine staining (ICS), that allows the enumeration of IFN-γ-producing TAA-specific CD8+ T cells in various tissues (tumor, blood, lymphoid organs) following oncolytic vaccination.
Collapse
Affiliation(s)
- Jonathan G Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France. .,INSERM, U1138, Paris, France. .,Equipe 11 Labellisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France. .,Université de Paris, Paris, France. .,Sorbonne Université, Paris, France.
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Brian D Lichty
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada. .,Turnstone Biologics, Ottawa, ON, Canada.
| |
Collapse
|
31
|
Rakshit S, Ahmed A, Adiga V, Sundararaj BK, Sahoo PN, Kenneth J, D’Souza G, Bonam W, Johnson C, Franken KL, Ottenhoff TH, Finak G, Gottardo R, Stuart KD, De Rosa SC, McElrath MJ, Vyakarnam A. BCG revaccination boosts adaptive polyfunctional Th1/Th17 and innate effectors in IGRA+ and IGRA- Indian adults. JCI Insight 2019; 4:130540. [PMID: 31743110 PMCID: PMC6975271 DOI: 10.1172/jci.insight.130540] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUNDBacille Calmette-Guérin (BCG) vaccine is protective against Tuberculosis (TB) in children, but its efficacy wanes with age. Consequently, determining if BCG revaccination augments anti-TB immunity in young adults in TB endemic regions is vital.METHODSTwo hundred healthy adults, BCG vaccinated at birth, were tested for their IFN-γ release assay (IGRA) status. Of these, 28 IGRA+ and 30 IGRA- were BCG revaccinated, and 24 IGRA+ and 23 IGRA- subjects served as unvaccinated controls. T and innate cell responses to mycobacterial antigens were analyzed by 14-color flow cytometry over 34 weeks.RESULTSIFN-γ and/or IL-2 Ag85A- and BCG-specific CD4+ and CD8+ T cell responses were boosted by revacciantion at 4 and 34 weeks, respectively, and were > 2-fold higher in IGRA+ compared with IGRA- vaccinees. Polyfunctional Ag85A, BCG, and mycobacterium tuberculosis (Mtb) latency Ag-specific (LTAg-specific) CD4+ T cells expressing up to 8 cytokines were also significantly enhanced in both IGRA+ and IGRA- vaccinees relative to unvaccinated controls, most markedly in IGRA+ vaccinees. A focused analysis of Th17 responses revealed expansion of Ag85A-, BCG-, and LTAg-specific total IL-17A+,IL-17F+,IL-22+, and IL-10+ CD4+ T cell effectors in both IGRA+ and IGRA- subjects. Also, innate IFN-γ+ NK/γδ/NKT cell responses were higher in both IGRA+ and IGRA- vaccinees compared with controls. This is the first evidence to our knowledge that BCG revaccination significantly boosts antimycobacterial Th1/Th17 responses in IGRA+ and IGRA- subjects.CONCLUSIONThese data show that BCG revaccination is immunogenic in IGRA- and IGRA+ subjects, implying that Mtb preinfection in IGRA+ subjects does not impact immunogenicity. This has implications for public health and vaccine development strategies.FUNDINGThis work was funded principally by DBT-NIH (BT/MB/Indo-US/HIPC/2013).
Collapse
Affiliation(s)
- Srabanti Rakshit
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Asma Ahmed
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Vasista Adiga
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Bharath K. Sundararaj
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Pravat Nalini Sahoo
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - John Kenneth
- Department of Infectious Diseases and
- Department of Pulmonary Medicine, St. John’s Research Institute, Bangalore, India
| | - George D’Souza
- Department of Infectious Diseases and
- Department of Pulmonary Medicine, St. John’s Research Institute, Bangalore, India
| | | | | | - Kees L.M.C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H.M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Greg Finak
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Laboratory Medicine and
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Annapurna Vyakarnam
- Laboratory of Immunology of HIV-TB Co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, Guy’s Hospital, King’s College London, London, United Kingdom
| |
Collapse
|
32
|
Singh M, Bhatt P, Sharma M, Varma-Basil M, Chaudhry A, Sharma S. Immunogenicity of late stage specific peptide antigens of Mycobacterium tuberculosis. INFECTION GENETICS AND EVOLUTION 2019; 74:103930. [PMID: 31228643 DOI: 10.1016/j.meegid.2019.103930] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/06/2019] [Accepted: 06/18/2019] [Indexed: 01/05/2023]
Abstract
Global burden of latent TB infection comprises one-third of the world population. Identifying potential Mycobacterium tuberculosis (Mtb) latency associated antigens that can generate protective immunity against the pathogen is crucial for designing an effective TB vaccine. Usually the immune system responds to a small number of amino acids as MHC Class I or Class II peptides. The precision to trigger epitope specific protective T-cell immune response could therefore be achieved with synthetic peptide-based subunit vaccine. In the present study we have considered an immunoinformatic approach using available softwares (ProPred, IEDB, NETMHC, BIMAS, Vaxijen2.0) and docking and visualizing softwares (CABSDOCK, HEX, Pymol, Discovery Studio) to select 10 peptides as latency antigens from 4 proteins (Rv2626, Rv2627, Rv2628, and Rv2032) of DosR regulon of Mtb. As Intracellular IFN-γ secreted by T cells is the most essential cytokine in Th1 mediated protective immunity, these peptides were verified as potential immunogenic epitopes in Peripheral Blood Mononuclear Cells (PBMCs) of 10 healthy contacts of TB patients (HTB) and 10 Category I Pulmonary TB patients (PTB).The antigen-specific CD4 and CD8 T cells expressing intracellular IFN-γ were analyzed using monoclonal antibodies in all subjects by multi-parameter flow cytometry. Both, PTB and HTB individuals responded to DosR peptides by showing increased frequency of IFN-γ+CD4 and IFN-γ+CD8 T cells. The T-cell responses were significantly higher in PTB patients in comparision to the HTB individuals. Additionally, our synthetic peptides and pools showed higher frequencies of IFN-γ+CD4 and IFN-γ+CD8 T cells than the peptides of Ag85B. This pilot study can be taken up further in larger sample size which may support the untapped opportunity of designing Mtb DosR inclusive peptide based post-exposure subunit vaccine.
Collapse
Affiliation(s)
- Medha Singh
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Parul Bhatt
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | - Monika Sharma
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India
| | | | - Anil Chaudhry
- Rajan Babu Institute of Pulmonary Medicine and Tuberculosis Hospital, GTB Nagar, Delhi 110009, India
| | - Sadhna Sharma
- DS Kothari Centre for Research and Innovation in Science Education, Miranda House and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| |
Collapse
|
33
|
Ramaiah A, Nayak S, Rakshit S, Manson AL, Abeel T, Shanmugam S, Sahoo PN, John AJUK, Sundaramurthi JC, Narayanan S, D'Souza G, von Hoegen P, Ottenhoff THM, Swaminathan S, Earl AM, Vyakarnam A. Evidence for Highly Variable, Region-Specific Patterns of T-Cell Epitope Mutations Accumulating in Mycobacterium tuberculosis Strains. Front Immunol 2019; 10:195. [PMID: 30814998 PMCID: PMC6381025 DOI: 10.3389/fimmu.2019.00195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/23/2019] [Indexed: 01/26/2023] Open
Abstract
Vaccines that confer protection through induction of adaptive T-cell immunity rely on understanding T-cell epitope (TCE) evolution induced by immune escape. This is poorly understood in tuberculosis (TB), an ancient, chronic disease, where CD4 T-cell immunity is of recognized importance. We probed 905 functionally validated, curated human CD4 T cell epitopes in 79 Mycobacterium tuberculosis (Mtb) whole genomes from India. This screen resulted in identifying 64 mutated epitopes in these strains initially using a computational pipeline and subsequently verified by single nucleotide polymorphism (SNP) analysis. SNP based phylogeny revealed the 79 Mtb strains to cluster to East African Indian (EAI), Central Asian Strain (CAS), and Beijing (BEI) lineages. Eighty-nine percent of the mutated T-cell epitopes (mTCEs) identified in the 79 Mtb strains from India has not previously been reported. These mTCEs were encoded by genes with high nucleotide diversity scores including seven mTCEs encoded by six antigens in the top 10% of rapidly divergent Mtb genes encoded by these strains. Using a T cell functional assay readout, we demonstrate 62% of mTCEs tested to significantly alter CD4 T-cell IFNγ and/or IL2 secretion with associated changes in predicted HLA-DR binding affinity: the gain of function mutations displayed higher predicted HLA-DR binding affinity and conversely mutations resulting in loss of function displayed lower predicted HLA-DR binding affinity. Most mutated antigens belonged to the cell wall/cell processes, and, intermediary metabolism and respiration families though all known Mtb proteins encoded mutations. Analysis of the mTCEs in an SNP database of 5,310 global Mtb strains identified 82% mTCEs to be significantly more prevalent in Mtb strains isolated from India, including 36 mTCEs identified exclusively in strains from India. These epitopes had a significantly higher predicted binding affinity to HLA-DR alleles that were highly prevalent in India compared to HLA-DR alleles rare in India, highlighting HLA-DR maybe an important driver of these mutations. This first evidence of region-specific TCE mutations potentially employed by Mtb to escape host immunity has important implications for TB vaccine design.
Collapse
Affiliation(s)
- Arunachalam Ramaiah
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Soumya Nayak
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Srabanti Rakshit
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Abigail L Manson
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Thomas Abeel
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | | | - Pravat Nalini Sahoo
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | | | | | - Sujatha Narayanan
- National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - George D'Souza
- Department of Pulmonary Medicine, St. John's Research Institute, Bangalore, India
| | | | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Ashlee M Earl
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Annapurna Vyakarnam
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India.,Department of Infectious Diseases, Faculty of Life Sciences & Medicine, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
34
|
Dey S, Kamil Reza K, Wuethrich A, Korbie D, Ibn Sina AA, Trau M. Tracking antigen specific T-cells: Technological advancement and limitations. Biotechnol Adv 2018; 37:145-153. [PMID: 30508573 DOI: 10.1016/j.biotechadv.2018.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/30/2018] [Accepted: 11/20/2018] [Indexed: 11/18/2022]
Abstract
Assessing T-cell mediated immune status can help to understand the body's response to disease and also provide essential diagnostic information. However, detection and characterization of immune response are challenging due to the rarity of signature biomolecules in biological fluid and require highly sensitive and specific assay technique for the analysis. Until now, several techniques spanning from flow cytometry to microsensors have been developed or under investigation for T-cell mediated immune response monitoring. Most of the current assays are designed to estimate average immune responses, i.e., total functional protein analysis and detection of total T-cells irrespective of their antigen specificity. Although potential, immune response analysis without detecting and characterizing the rare subset of T-cell population could lead to over or underestimation of patient's immune status. Addressing this limitation, recently a number of technological advancements in biosensing have been developed for this. The potential of simple and precise micro-technologies including microarray and microfluidic platforms for assessing antigen-specific T-cells will be highlighted in this review, together with a discussion on existing challenges and future aspects of immune-sensor development.
Collapse
Affiliation(s)
- Shuvashis Dey
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - K Kamil Reza
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Alain Wuethrich
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Darren Korbie
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia
| | - Abu Ali Ibn Sina
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia.
| | - Matt Trau
- Centre for Personalised Nanomedicine, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, QLD 4072, Australia; School of Chemistry and Molecular Biosciences, The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
35
|
Bowyer G, Rampling T, Powlson J, Morter R, Wright D, Hill AVS, Ewer KJ. Activation-induced Markers Detect Vaccine-Specific CD4⁺ T Cell Responses Not Measured by Assays Conventionally Used in Clinical Trials. Vaccines (Basel) 2018; 6:vaccines6030050. [PMID: 30065162 PMCID: PMC6161310 DOI: 10.3390/vaccines6030050] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 12/27/2022] Open
Abstract
Immunogenicity of T cell-inducing vaccines, such as viral vectors or DNA vaccines and Bacillus Calmette-Guérin (BCG), are frequently assessed by cytokine-based approaches. While these are sensitive methods that have shown correlates of protection in various vaccine studies, they only identify a small proportion of the vaccine-specific T cell response. Responses to vaccination are likely to be heterogeneous, particularly when comparing prime and boost or assessing vaccine performance across diverse populations. Activation-induced markers (AIM) can provide a broader view of the total antigen-specific T cell response to enable a more comprehensive evaluation of vaccine immunogenicity. We tested an AIM assay for the detection of vaccine-specific CD4+ and CD8+ T cell responses in healthy UK adults vaccinated with viral vectored Ebola vaccine candidates, ChAd3-EBO-Z and MVA-EBO-Z. We used the markers, CD25, CD134 (OX40), CD274 (PDL1), and CD107a, to sensitively identify vaccine-responsive T cells. We compared the use of OX40+CD25+ and OX40+PDL1+ in CD4+ T cells and OX40+CD25+ and CD25+CD107a+ in CD8+ T cells for their sensitivity, specificity, and associations with other measures of vaccine immunogenicity. We show that activation-induced markers can be used as an additional method of demonstrating vaccine immunogenicity, providing a broader picture of the global T cell response to vaccination.
Collapse
Affiliation(s)
- Georgina Bowyer
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Tommy Rampling
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | | | - Richard Morter
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Daniel Wright
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
36
|
Abstract
Tuberculosis is a complex disease, which can affect many organs other than the lungs. Initial infection may be cleared without inducing immunological memory, or progress directly to primary disease. Alternatively, the infection may be controlled as latent TB infection, that may progress to active tuberculosis at a later stage. There is now a greater understanding that these infection states are part of a continuum, and studies using PET/CT imaging have shown that individual lung granulomas may respond to infection independently, in an un-synchronized manner. In addition, the Mycobacterium tuberculosis organisms themselves can exist in different states: as nonculturable forms, as 'persisters', as rapidly growing bacteria and a biofilm-forming cording phenotype. The 'omics' approaches of transcriptomics, metabolomics and proteomics can help reveal the mechanisms underlying these different infection states in the host, and identify biosignatures with diagnostic potential, that can predict the development of disease, in 'progressors' as early as 12-18 months before it can be detected clinically, or that can monitor the success of anti-TB therapy. Further insights can be obtained from studies of BCG vaccination and new TB vaccines. For example, epigenetic changes associated with trained immunity and a stronger immune responses following BCG vaccination can be identified. These omics approaches may be particularly valuable when linked to studies of mycobacterial growth inhibition, as a direct read-out of the ability to control mycobacterial growth. The second generation of omics studies is identifying much smaller signatures based on as few as 3 or 4 genes. Thus, narrowing down omics-derived biosignatures to a manageable set of markers now opens the way to field-friendly point of care assays.
Collapse
Affiliation(s)
- M Lerm
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - H M Dockrell
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
37
|
New Technologies for Vaccine Development: Harnessing the Power of Human Immunology. J Indian Inst Sci 2018. [DOI: 10.1007/s41745-018-0064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
38
|
Baumgaertner P, Ioannidou K, Speiser DE. Immune Monitoring of Blood and Tumor Microenvironment. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
39
|
Biomarker-guided stratification of autoimmune patients for biologic therapy. Curr Opin Immunol 2017; 49:56-63. [DOI: 10.1016/j.coi.2017.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/22/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023]
|
40
|
Assay optimisation and technology transfer for multi-site immuno-monitoring in vaccine trials. PLoS One 2017; 12:e0184391. [PMID: 29020010 PMCID: PMC5636064 DOI: 10.1371/journal.pone.0184391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/23/2017] [Indexed: 11/28/2022] Open
Abstract
Cellular immunological assays are important tools for the monitoring of responses to T-cell-inducing vaccine candidates. As these bioassays are often technically complex and require considerable experience, careful technology transfer between laboratories is critical if high quality, reproducible data that allows comparison between sites, is to be generated. The aim of this study, funded by the European Union Framework Program 7-funded TRANSVAC project, was to optimise Standard Operating Procedures and the technology transfer process to maximise the reproducibility of three bioassays for interferon-gamma responses: enzyme-linked immunosorbent assay (ELISA), ex-vivo enzyme-linked immunospot and intracellular cytokine staining. We found that the initial variability in results generated across three different laboratories reduced following a combination of Standard Operating Procedure harmonisation and the undertaking of side-by-side training sessions in which assay operators performed each assay in the presence of an assay ‘lead’ operator. Mean inter-site coefficients of variance reduced following this training session when compared with the pre-training values, most notably for the ELISA assay. There was a trend for increased inter-site variability at lower response magnitudes for the ELISA and intracellular cytokine staining assays. In conclusion, we recommend that on-site operator training is an essential component of the assay technology transfer process and combined with harmonised Standard Operating Procedures will improve the quality, reproducibility and comparability of data produced across different laboratories. These data may be helpful in ongoing discussions of the potential risk/benefit of centralised immunological assay strategies for large clinical trials versus decentralised units.
Collapse
|
41
|
Kaufmann SHE, Dockrell HM, Drager N, Ho MM, McShane H, Neyrolles O, Ottenhoff THM, Patel B, Roordink D, Spertini F, Stenger S, Thole J, Verreck FAW, Williams A. TBVAC2020: Advancing Tuberculosis Vaccines from Discovery to Clinical Development. Front Immunol 2017; 8:1203. [PMID: 29046674 PMCID: PMC5632681 DOI: 10.3389/fimmu.2017.01203] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/11/2017] [Indexed: 01/24/2023] Open
Abstract
TBVAC2020 is a research project supported by the Horizon 2020 program of the European Commission (EC). It aims at the discovery and development of novel tuberculosis (TB) vaccines from preclinical research projects to early clinical assessment. The project builds on previous collaborations from 1998 onwards funded through the EC framework programs FP5, FP6, and FP7. It has succeeded in attracting new partners from outstanding laboratories from all over the world, now totaling 40 institutions. Next to the development of novel vaccines, TB biomarker development is also considered an important asset to facilitate rational vaccine selection and development. In addition, TBVAC2020 offers portfolio management that provides selection criteria for entry, gating, and priority settings of novel vaccines at an early developmental stage. The TBVAC2020 consortium coordinated by TBVI facilitates collaboration and early data sharing between partners with the common aim of working toward the development of an effective TB vaccine. Close links with funders and other consortia with shared interests further contribute to this goal.
Collapse
Affiliation(s)
- Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hazel M Dockrell
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nick Drager
- Tuberculosis Vaccine Initiative (TBVI), Lelystad, Netherlands
| | - Mei Mei Ho
- Bacteriology Division, MHRA-NIBSC, Potters Bar, United Kingdom
| | | | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Brij Patel
- RegExcel Consulting Ltd, Surrey, United Kingdom
| | | | | | | | - Jelle Thole
- Tuberculosis Vaccine Initiative (TBVI), Lelystad, Netherlands
| | | | | | | |
Collapse
|
42
|
Titball RW, Burtnick MN, Bancroft GJ, Brett P. Burkholderia pseudomallei and Burkholderia mallei vaccines: Are we close to clinical trials? Vaccine 2017; 35:5981-5989. [DOI: 10.1016/j.vaccine.2017.03.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/17/2017] [Accepted: 03/07/2017] [Indexed: 10/19/2022]
|
43
|
Circulating Mycobacterium tuberculosis DosR latency antigen-specific, polyfunctional, regulatory IL10 + Th17 CD4 T-cells differentiate latent from active tuberculosis. Sci Rep 2017; 7:11948. [PMID: 28931830 PMCID: PMC5607261 DOI: 10.1038/s41598-017-10773-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/02/2017] [Indexed: 12/30/2022] Open
Abstract
The functional heterogeneity of T cell responses to diverse antigens expressed at different stages of Mycobacterium tuberculosis (Mtb) infection, in particular early secreted versus dormancy related latency antigens expressed later, that distinguish subjects with latent (LTBI), pulmonary (PTB) or extrapulmonary (EPTB) tuberculosis remains unclear. Here we show blood central memory CD4 T-cell responses specific to Mtb dormancy related (DosR) latency, but not classical immunodominant secretory antigens, to clearly differentiate LTBI from EPTB and PTB. The polyfunctionality score integrating up to 31 DosR-specific CD4 T-cell functional profiles was significantly higher in LTBI than EPTB or PTB subjects. Further analysis of 256 DosR-specific T-cell functional profiles identified regulatory IL10 + Th17 cells (IL10+IL17A+IL17F+IL22+) to be significantly enriched in LTBI; in contrast to pro-inflammatory Th17 cells (IFNγ+IL17A+/IL10-) in the blood and lung of EPTB and PTB subjects respectively. A blood polyfunctional, Mtb DosR latency antigen specific, regulatory, central memory response is therefore a novel functional component of T-cell immunity in latent TB and potential correlate of protection.
Collapse
|
44
|
Dreesman A, Corbière V, Dirix V, Smits K, Debulpaep S, De Schutter I, Libin M, Singh M, Malfroot A, Locht C, Mascart F. Age-Stratified T Cell Responses in Children Infected with Mycobacterium tuberculosis. Front Immunol 2017; 8:1059. [PMID: 28928738 PMCID: PMC5591888 DOI: 10.3389/fimmu.2017.01059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/15/2017] [Indexed: 12/19/2022] Open
Abstract
Tuberculosis (TB) in young children differs from adult TB in that the risk of rapid progression to active TB (aTB) is higher in children than in adults. The reasons for this increased risk are not fully understood. Early differentiation remains difficult between children at risk to develop aTB from those who will remain healthy and develop a latent TB infection (LTBI). Biomarkers to differentiate aTB from LTBI in children, especially in very young children, are urgently needed. To identify M. tuberculosis-specific functional T cell subsets related to clinical manifestations in children, we enrolled 87 children exposed to M. tuberculosis. After standard clinical assessment, the children were classified as aTB, LTBI, or uninfected. Their CD4+ T cell cytokine profiles (IFN-γ, TNF-α, IL-2, IL-17) were analyzed at the single-cell level by flow cytometry after stimulation with three mycobacterial antigens, purified protein derivative (PPD), early-secreted-antigenic target-6 (ESAT-6), or heparin-binding hemagglutinin (HBHA). This approach identified age-related discriminative markers between aTB and LTBI. Whereas among the 3- to 15-year-old children, an excellent discrimination between aTB and LTBI was provided by comparing the ratio between the proportions of ESAT-6-induced IFN-γsingle+ and ESAT-6-induced TNF-αsingle+CD4+ T lymphocytes, this was not the case for children younger than 3 years. By contrast, in this group (<3years), the analysis of HBHA-induced IL-17single+CD4+ T lymphocytes allowed us to identify children with LTBI by the high proportion of this cellular lymphocyte subset, whereas this was not the case for children with aTB. The analysis at the single-cell level of T cell immune responses induced by mycobacterial antigens are, thus, different in infected children younger or older than 3 years of age. HBHA-induced IL-17 production by CD4+ T lymphocytes was associated with protection only in children under 3 years who are at high risk for rapid progression to aTB. This suggests that the HBHA-induced IL-17 production by CD4+ T lymphocytes is a potential new correlate of protection against M. tuberculosis in humans, and that the distinction between children with LTBI and those with aTB is possible based on age-related diagnostic markers.
Collapse
Affiliation(s)
- Alexandra Dreesman
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Véronique Corbière
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Violette Dirix
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Kaat Smits
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Sara Debulpaep
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium.,Department of Pediatrics, CHU Saint-Pierre, Brussels, Belgium
| | - Iris De Schutter
- Department of Pediatric Pulmonology, Cystic Fibrosis Clinic and Pediatric Infectious Diseases, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Myriam Libin
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Mahavir Singh
- Lionex Diagnostics and Therapeutics, Braunschweig, Germany
| | - Anne Malfroot
- Department of Pediatric Pulmonology, Cystic Fibrosis Clinic and Pediatric Infectious Diseases, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Camille Locht
- INSERM, U1019, Lille, France.,CNRS, UMR8204, Lille, France.,Université de Lille, Lille, France.,Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Françoise Mascart
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium.,Immunobiology Clinic, Hôpital Erasme, Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| |
Collapse
|
45
|
Preferential Targeting of Conserved Gag Regions after Vaccination with a Heterologous DNA Prime-Modified Vaccinia Virus Ankara Boost HIV-1 Vaccine Regimen. J Virol 2017; 91:JVI.00730-17. [PMID: 28701395 DOI: 10.1128/jvi.00730-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/14/2017] [Indexed: 11/20/2022] Open
Abstract
Prime-boost vaccination strategies against HIV-1 often include multiple variants for a given immunogen for better coverage of the extensive viral diversity. To study the immunologic effects of this approach, we characterized breadth, phenotype, function, and specificity of Gag-specific T cells induced by a DNA-prime modified vaccinia virus Ankara (MVA)-boost vaccination strategy, which uses mismatched Gag immunogens in the TamoVac 01 phase IIa trial. Healthy Tanzanian volunteers received three injections of the DNA-SMI vaccine encoding a subtype B and AB-recombinant Gagp37 and two vaccinations with MVA-CMDR encoding subtype A Gagp55 Gag-specific T-cell responses were studied in 42 vaccinees using fresh peripheral blood mononuclear cells. After the first MVA-CMDR boost, vaccine-induced gamma interferon-positive (IFN-γ+) Gag-specific T-cell responses were dominated by CD4+ T cells (P < 0.001 compared to CD8+ T cells) that coexpressed interleukin-2 (IL-2) (66.4%) and/or tumor necrosis factor alpha (TNF-α) (63.7%). A median of 3 antigenic regions were targeted with a higher-magnitude median response to Gagp24 regions, more conserved between prime and boost, compared to those of regions within Gagp15 (not primed) and Gagp17 (less conserved; P < 0.0001 for both). Four regions within Gagp24 each were targeted by 45% to 74% of vaccinees upon restimulation with DNA-SMI-Gag matched peptides. The response rate to individual antigenic regions correlated with the sequence homology between the MVA- and DNA Gag-encoded immunogens (P = 0.04, r2 = 0.47). In summary, after the first MVA-CMDR boost, the sequence-mismatched DNA-prime MVA-boost vaccine strategy induced a Gag-specific T-cell response that was dominated by polyfunctional CD4+ T cells and that targeted multiple antigenic regions within the conserved Gagp24 protein.IMPORTANCE Genetic diversity is a major challenge for the design of vaccines against variable viruses. While including multiple variants for a given immunogen in prime-boost vaccination strategies is one approach that aims to improve coverage for global virus variants, the immunologic consequences of this strategy have been poorly defined so far. It is unclear whether inclusion of multiple variants in prime-boost vaccination strategies improves recognition of variant viruses by T cells and by which mechanisms this would be achieved, either by improved cross-recognition of multiple variants for a given antigenic region or through preferential targeting of antigenic regions more conserved between prime and boost. Engineering vaccines to induce adaptive immune responses that preferentially target conserved antigenic regions of viral vulnerability might facilitate better immune control after preventive and therapeutic vaccination for HIV and for other variable viruses.
Collapse
|
46
|
Elnaggar MM, Abdellrazeq GS, Elsisy A, Mahmoud AH, Shyboub A, Sester M, Khaliel SA, Singh M, Torky HA, Davis WC. Evaluation of antigen specific interleukin-1β as a biomarker to detect cattle infected with Mycobacterium bovis. Tuberculosis (Edinb) 2017; 105:53-59. [PMID: 28610788 DOI: 10.1016/j.tube.2017.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/04/2017] [Accepted: 04/21/2017] [Indexed: 01/01/2023]
Abstract
Bovine tuberculosis (bTB) is a major world-wide health problem that has been difficult to control, due to the lack of an effective vaccine and limited ability of the tuberculin skin test (TST) and the ancillary whole blood interferon-gamma (IFN-γ) release assay (IGRA) to detect all infected animals. A 6 h cytokine flow cytometric IFN-γ (CFC) assay was developed in effort to overcome these limitations and expand methods for studying the mechanisms of bTB immunopathogenesis. The present study was conducted to evaluate IL-1β as a biomarker to use in conjunction with the IFN-γ CFC assay to improve the diagnostic accuracy for bTB. Three animal groups with predefined Mbv infection status were used for analysis of IL-1β in plasma from whole blood cultures stimulated with ESAT-6/CFP-10 for 20-24 h. Parallel stimulations were performed for enumeration of IFN-γ producing T cells. Data analysis showed that Mbv infected animals have a higher frequency of IFN-γ producing CD4+ T cells and plasma IL-1β than animals exposed to non-tuberculous mycobacteria (NTM) or uninfected control animals, with a significant correlation between the two readouts, thus allowing differentiation between the three animal groups. IL-1β has the potential to serve as an additional biomarker for detecting cattle infected with Mbv.
Collapse
Affiliation(s)
- Mahmoud M Elnaggar
- Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt; Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Pullman, WA, USA.
| | - Gaber S Abdellrazeq
- Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt; Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Pullman, WA, USA
| | - Alaa Elsisy
- Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt
| | - Asmaa H Mahmoud
- Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt
| | - Abdelrazeq Shyboub
- Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt
| | - Martina Sester
- Department of Transplant and Infection Immunology, Institutes for Infection Medicine, Saarland University, Homburg, Germany
| | - Samy A Khaliel
- Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt
| | - Mahavir Singh
- Lionex Diagnostics & Therapeutics, Braunschweig, Germany
| | - Helmy A Torky
- Department of Microbiology, Faculty of Veterinary Medicine, Alexandria University, Egypt
| | - William C Davis
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Pullman, WA, USA
| |
Collapse
|
47
|
Wowk PF, Franco LH, Fonseca DMD, Paula MO, Vianna ÉDSO, Wendling AP, Augusto VM, Elói-Santos SM, Teixeira-Carvalho A, Silva FDC, Vinhas SA, Martins-Filho OA, Palaci M, Silva CL, Bonato VLD. Mycobacterial Hsp65 antigen upregulates the cellular immune response of healthy individuals compared with tuberculosis patients. Hum Vaccin Immunother 2017; 13:1040-1050. [PMID: 28059670 DOI: 10.1080/21645515.2016.1264547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Previously we showed that 65-kDa Mycobacterium leprae heat shock protein (Hsp65) is a target for the development of a tuberculosis vaccine. Here we evaluated peripheral blood mononuclear cells (PBMC) from healthy individuals or tuberculosis patients stimulated with two forms of Hsp65 antigen, recombinant DNA that encodes Hsp65 (DNA-HSP65) or recombinant Hsp65 protein (rHsp65) in attempting to mimic a prophylactic or therapeutic study in vitro, respectively. Proliferation and cytokine-producing CD4+ or CD8+ cell were assessed by flow cytometry. The CD4+ cell proliferation from healthy individuals was stimulated by DNA-HSP65 and rHsp65, while CD8+ cell proliferation from healthy individuals or tuberculosis patients was stimulated by rHSP65. DNA-HSP65 did not improve the frequency of IFN-gamma+ cells from healthy individuals or tuberculosis patients. Furthermore, we found an increase in the frequency of IL-10-producing cells in both groups. These findings show that Hsp65 antigen activates human lymphocytes and plays an immune regulatory role that should be addressed as an additional antigen for the development of antigen-combined therapies.
Collapse
Affiliation(s)
- Pryscilla Fanini Wowk
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Luís Henrique Franco
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Denise Morais da Fonseca
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Marina Oliveira Paula
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | | | - Ana Paula Wendling
- c Laboratory of Biomarkers for Diagnosis and Monitoring , René Rachou Research Center , Fiocruz, Belo Horizonte , Minas Gerais , Brazil
| | | | | | - Andréa Teixeira-Carvalho
- c Laboratory of Biomarkers for Diagnosis and Monitoring , René Rachou Research Center , Fiocruz, Belo Horizonte , Minas Gerais , Brazil
| | - Flávia Dias Coelho Silva
- e Mycobacteriology Laboratory, Nucleus of Infectious Diseases , Federal University of Espírito Santo , Vitória , Espírito Santos , Brazil
| | - Solange Alves Vinhas
- e Mycobacteriology Laboratory, Nucleus of Infectious Diseases , Federal University of Espírito Santo , Vitória , Espírito Santos , Brazil
| | - Olindo Assis Martins-Filho
- c Laboratory of Biomarkers for Diagnosis and Monitoring , René Rachou Research Center , Fiocruz, Belo Horizonte , Minas Gerais , Brazil
| | - Moisés Palaci
- e Mycobacteriology Laboratory, Nucleus of Infectious Diseases , Federal University of Espírito Santo , Vitória , Espírito Santos , Brazil
| | - Célio Lopes Silva
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| | - Vânia Luiza Deperon Bonato
- a Department of Biochemistry and Immunology , Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto , Brazil
| |
Collapse
|
48
|
Abstract
New and effective tuberculosis (TB) vaccines are urgently needed to control pulmonary TB, and in particular to prevent the spread of drug-resistant strains of Mycobacterium tuberculosis. These drug-resistant strains can range from those resistant to first-line drugs to those that are almost impossible to treat. To develop new and effective vaccines for HIV and malaria has been difficult and it is proving to be just as challenging for TB. TB is a complicated disease with a spectrum from apparently controlled latent infection to active clinical disease and so different types of preventive or post-exposure vaccine may be needed. Identifying the most promising vaccine candidates to move into clinical trials is difficult, as we lack biomarker signatures that can predict protective efficacy. There is a risk that the failure of the MVA-85A vaccine to show efficacy when given to previously BCG-vaccinated South African infants will impact on the resources available for the development and trials of other candidate TB vaccines. Continued support for the development of new TB vaccines should remain a priority as an effective vaccine would bring huge public health benefits.
Collapse
|
49
|
Nemes E, Burgers WA, Riou C, Andersen-Nissen E, Ferrari G, Gray CM, Scriba T. Teaching advanced flow cytometry in Africa: 10 years of lessons learned. Cytometry A 2016; 89:971-974. [PMID: 27870536 DOI: 10.1002/cyto.a.23015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/03/2016] [Accepted: 10/08/2016] [Indexed: 01/24/2023]
Affiliation(s)
- Elisa Nemes
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, South Africa.,Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Wendy A Burgers
- Division of Medical Virology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Catherine Riou
- Division of Medical Virology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Erica Andersen-Nissen
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Guido Ferrari
- Department of Surgery, Duke University, Durham, North Carolina
| | - Clive M Gray
- Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Thomas Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), University of Cape Town, South Africa.,Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| |
Collapse
|
50
|
Multifunctional T Cell Response to DosR and Rpf Antigens Is Associated with Protection in Long-Term Mycobacterium tuberculosis-Infected Individuals in Colombia. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:813-824. [PMID: 27489136 DOI: 10.1128/cvi.00217-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/27/2016] [Indexed: 01/03/2023]
Abstract
Multifunctional T cells have been shown to be protective in chronic viral infections. In mycobacterial infections, however, evidence for a protective role of multifunctional T cells remains inconclusive. Short-term cultures of peripheral blood mononuclear cells stimulated with the Mycobacterium tuberculosis RD1 antigens 6-kDa early secretory antigenic target (ESAT6) and 10-kDa culture filtrate antigen (CFP10), which are induced in the early infection phase, have been mainly used to assess T cell multifunctionality, although long-term culture assays have been proposed to be more sensitive than short-term assays for assessment of memory T cells, which are essential for long-term immunity. Here we used a long-term culture assay system to study the T cell immune responses to the M. tuberculosis latency-associated DosR antigens and reactivation-associated Rpf antigens, compared to ESAT6 and CFP10, in patients with pulmonary tuberculosis (PTB) and household contacts of PTB patients with long-term latent tuberculosis infection (ltLTBI), in a community in which M. tuberculosis is endemic. Our results showed that the DosR antigens Rv1737c (narK2) and Rv2029c (pfkB) and the Rv2389c (rpfD) antigen of M. tuberculosis induced higher frequencies of CD4+ or CD8+ mono- or bifunctional (but not multifunctional) T cells producing interferon gamma (IFN-γ) and/or tumor necrosis alpha (TNF-α) in ltLTBI, compared to PTB. Moreover, the frequencies of CD4+ and/or CD8+ T cells with a CD45RO+ CD27+ phenotype were higher in ltLTBI than in PTB. Thus, the immune responses to selected DosR and Rpf antigens may be associated with long-term latency, correlating with protection from M. tuberculosis reactivation in ltLTBI. Further study of the functional and memory phenotypes may contribute to further discrimination between the different states of M. tuberculosis infections.
Collapse
|