1
|
Li Y, Ashuo A, Hao M, Li Y, Ye J, Liu J, Hua T, Fang Z, Li J, Yuan Z, Chen J. An extracellular humanized IFNAR immunocompetent mouse model for analyses of human interferon alpha and subtypes. Emerg Microbes Infect 2024; 13:2287681. [PMID: 37994664 PMCID: PMC10810641 DOI: 10.1080/22221751.2023.2287681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023]
Abstract
Type I interferons (IFN-Is) have key roles in immune defense and treatments for various diseases, including chronic hepatitis B virus (HBV) infection. All IFN-Is signal through a shared IFN-I heterodimeric receptor complex comprising IFN-α receptor 1 (IFNAR1) and IFNAR2 subunits, but differences in antiviral and immunomodulatory responses among IFN-I subtypes remain largely unknown. Because the IFN-IFNAR interactions are species-specific, mice exhibit weak responses to human IFN-I. To more fully characterize the actions of human IFN-α and its subtypes in vivo, a gene targeting strategy was employed to generate gene knock-in mice with extracellular-humanized IFNAR1/2 (IFNAR-hEC) in the C57BL/6N strain. IFNAR-hEC mice actively responded to human IFN-I, and endogenous mouse IFN-I signalling remained active in heterozygous mice (IfnarhEC/+). Analyses of IFNAR-hEC mice and isolated cells showed that human IFN-α2 and α14 subtypes exerted differential effect on the activation of JAK-STAT signalling and immune responses. Compared with IFN-α2, IFN-α14 induced greater activation of STAT1/2 and IFN-stimulated genes, synergistically elicited IFN-α and -γ signalling, and induced higher numbers of antigen-specific CD8+ T cells. Moreover, IFNAR-hEC mice with HBV replication displayed long-term viral suppression upon treatment with the clinically-used PEGylated hIFN-α2. These results indicate that IFNAR-hEC mice may be useful for elucidating antiviral and immunomodulatory functions of human IFN-Is and for conducting preclinical studies. A better understanding of the distinct activities of IFN-α subtypes can provide insights concerning the development of improved IFN-based therapy.
Collapse
Affiliation(s)
- Yumeng Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Asha Ashuo
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Menghan Hao
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, People’s Republic of China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Jianyu Ye
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Jiangxia Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Ting Hua
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Zhong Fang
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Jianhua Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, People’s Republic of China
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
2
|
He Y, Liu X, Zhu F, Dai Z, Li X, Li L, Zhao B, Yuan H, Lu Y, Liang Z, Zhang Y, Zhang L. Single-Cell Secretome Profiling Enabled by Selective Enrichment and NanoLC-MS/MS Analysis. Angew Chem Int Ed Engl 2024:e202417351. [PMID: 39609103 DOI: 10.1002/anie.202417351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 11/30/2024]
Abstract
Recent advances in single-cell proteomics enable the direct profiling of thousands of proteins from a single mammalian cell. However, due to the bottlenecks in detecting low-abundance secreted proteins and extracellular vesicle (EV) proteins (collectively referred to as the secretome) against a background of high-abundance proteins in serum-containing culture medium, the comprehensive investigation of the secretome at the single-cell level using nanoLC-MS/MS still remains challenging. Herein, we report a novel single-cell secretome profiling (SCSP) method by integrating the metabolic labeling of newly synthesized proteins, click chemistry-based enrichment, and in situ digestion of the labeled secretome in an alkyne-functionalized capillary micro-reactor, followed by nanoLC-MS/MS analysis. By this method, an average of 389 protein groups were quantified from the secretome of single HeLa cells (n=17), with a total of 752 protein groups confidently identified in the single-cell secretome, which is a significant increase compared to the previously reported targeted analysis limited to dozens of secreted proteins by antibody recognition. These results indicated that our developed SCSP method would provide a powerful tool to gain insights into secretion heterogeneity and intercellular communication at the single-cell level.
Collapse
Affiliation(s)
- Yingyun He
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinxin Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Fengjiao Zhu
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhongpeng Dai
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Xiao Li
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Linmei Li
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Baofeng Zhao
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Huiming Yuan
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yao Lu
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhen Liang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Yukui Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lihua Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| |
Collapse
|
3
|
Wambacq WA, Apper E, Le Bourgot C, Barbe F, Lyu Y, Pelst M, Broeckx BJG, Devriendt B, Cox E, Hesta M. A new combination of a prebiotic and postbiotic mitigates immunosenescence in vaccinated healthy senior dogs. Front Vet Sci 2024; 11:1392985. [PMID: 39634761 PMCID: PMC11616177 DOI: 10.3389/fvets.2024.1392985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/18/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Elderly dogs often suffer from chronic diseases, in part attributed to immunosenescence, characterized by reduced blood CD4+ T cells (helper T cells) and elevation in the CD8+ T cells subset (cytotoxic T cells). Studies conducted in adult dogs suggested that supplementing short chain fructo-oligosaccharides (scFOS) or postbiotics derived from yeasts may positively influence the immune response. The aim of the current study was to investigate whether dietary supplementation with a combination of scFOS with a new yeast postbiotic (Profeed ADVANCED® called scFOS+) could have a positive influence on the immune status of senior dogs subjected to an immune challenge (Lyme disease vaccination). Methods To this end, 22 healthy senior client-owned dogs were divided into two groups: one group received a placebo diet without scFOS+ and the other group the basal diet supplemented with 1.1% scFOS+. In order to assess immune function, complete blood count, serum acute phase proteins, immunoglobulins, cytokines, T-cell subsets and antibody secreting cells were analyzed. Furthermore, faecal score and pH were recorded. Results and discussion Dogs fed the scFOS+ supplement had decreased total serum IgA concentrations (p < 0.01), which might suggest a more local IgA response in agreement with what was previously found when adult dogs were supplemented with β-1, 3/1, 6-glucan, a yeast-based product. More importantly, the present study demonstrated that feeding 1.1% scFOS+ to healthy senior dogs increased the CD4+:CD8+ T-cell (Helper:Cytotoxic T cell) ratio (p < 0.001) during and after vaccination against Lyme disease. Combining scFOS and yeast-derived postbiotics in the diet can therefore counter certain characteristics of T-cell immunosenescence in dogs.
Collapse
Affiliation(s)
- Wendy A. Wambacq
- Equine and Companion Animal Nutrition, Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | - Cindy Le Bourgot
- Scientific and Regulatory Affairs Department, Tereos, Moussy-le-Vieux, France
| | | | - Yang Lyu
- Equine and Companion Animal Nutrition, Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Michael Pelst
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bart J. G. Broeckx
- Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eric Cox
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Myriam Hesta
- Equine and Companion Animal Nutrition, Department of Morphology, Imaging, Orthopedics, Rehabilitation and Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
4
|
Wu Q, Pan X, Han D, Ma Z, Zhang H. New Insights into the Epidemiological Characteristics of Mycoplasma pneumoniae Infection before and after the COVID-19 Pandemic. Microorganisms 2024; 12:2019. [PMID: 39458327 PMCID: PMC11509874 DOI: 10.3390/microorganisms12102019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Mycoplasma pneumoniae (M. pneumoniae), a prevalent respiratory pathogen affecting children and adolescents, is known to trigger periodic global epidemics. The most recent significant outbreak commenced in the first half of 2023 and reached its peak globally during the autumn and winter months. Considering the worldwide repercussions of the COVID-19 pandemic, it has become increasingly essential to delve into the epidemiological characteristics of M. pneumoniae both before and after the pandemic. This review aims to provide a comprehensive analysis of the key features of M. pneumoniae epidemics in the pre-and post-COVID-19 contexts, including but not limited to shifts in the susceptible population, the molecular genotypes of the pathogen, the clinical manifestations, and potential new trends in drug resistance. Additionally, we will introduce the latest advancements in the diagnosis of M. pneumoniae.
Collapse
Affiliation(s)
- Qianyue Wu
- Clinical Lab in Children’s Hospital of Shanghai, Children’s Hospital of Shanghai Jiao Tong University, Shanghai 200040, China; (Q.W.); (X.P.); (D.H.); (Z.M.)
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| | - Xiaozhou Pan
- Clinical Lab in Children’s Hospital of Shanghai, Children’s Hospital of Shanghai Jiao Tong University, Shanghai 200040, China; (Q.W.); (X.P.); (D.H.); (Z.M.)
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| | - Dingding Han
- Clinical Lab in Children’s Hospital of Shanghai, Children’s Hospital of Shanghai Jiao Tong University, Shanghai 200040, China; (Q.W.); (X.P.); (D.H.); (Z.M.)
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| | - Zhan Ma
- Clinical Lab in Children’s Hospital of Shanghai, Children’s Hospital of Shanghai Jiao Tong University, Shanghai 200040, China; (Q.W.); (X.P.); (D.H.); (Z.M.)
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| | - Hong Zhang
- Clinical Lab in Children’s Hospital of Shanghai, Children’s Hospital of Shanghai Jiao Tong University, Shanghai 200040, China; (Q.W.); (X.P.); (D.H.); (Z.M.)
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| |
Collapse
|
5
|
Phelps A, Pazos-Castro D, Urselli F, Grydziuszko E, Mann-Delany O, Fang A, Walker TD, Guruge RT, Tome-Amat J, Diaz-Perales A, Waserman S, Boonyaratanakornkit J, Jordana M, Taylor JJ, Koenig JFE. Production and use of antigen tetramers to study antigen-specific B cells. Nat Protoc 2024; 19:727-751. [PMID: 38243093 DOI: 10.1038/s41596-023-00930-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/20/2023] [Indexed: 01/21/2024]
Abstract
B cells generate antibodies that provide protection from infection, but also cause pathology in autoimmune and allergic conditions. Antigen-specific B cells can be detected by binding their surface antibody receptors with native antigens conjugated to fluorescent probes, a technique that has revealed substantial insight into B cell activation and function. This protocol describes the process of generating fluorescent antigen tetramer probes and delineates a process of enriching large samples based on antigen-specificity for high-resolution analyses of the antigen-specific B cell repertoire. Enrichment of tetramer-binding cells allows for detection of antigen-specific B cells as rare as 1 in 100 million cells, providing sufficient resolution to study naive B cells and IgE-expressing cells by flow cytometry. The generation of antigen tetramers involves antigen biotinylation, assessment of biotin:antigen ratio for optimal tetramer loading and polymerization around a streptavidin-fluorophore backbone. We also describe the construction of a control tetramer to exclude B cells binding to the tetramer backbone. We provide a framework to validate whether tetramer probes are detecting true antigen-specific B cells and discuss considerations for experimental design. This protocol can be performed by researchers trained in basic biomedical/immunological research techniques, using instrumentation commonly found in most laboratories. Constructing the antigen and control tetramers takes 9 h, though their specificity should be assessed before experimentation and may take weeks to months depending on the method of validation. Sample enrichment requires ~2 h but is generally time and cost neutral as fewer cells are run through the flow cytometer.
Collapse
Affiliation(s)
- Allyssa Phelps
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Diego Pazos-Castro
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Centre for Plant Biotechnology and Genomics, Universidad Politécnica de Madrid - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria/Consejo Superior de Investigaciones Científicas (UPM-INIA/CSIC), Universidad Politécnica de Madrid, Madrid, Spain
- Department of Biotechnology-Plant Biology, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas (ETSIAAB), Universidad Politécnica de Madrid, Madrid, Spain
| | - Francesca Urselli
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Emily Grydziuszko
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Olivia Mann-Delany
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Allison Fang
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Tina D Walker
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Rangana Talpe Guruge
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Jaime Tome-Amat
- Centre for Plant Biotechnology and Genomics, Universidad Politécnica de Madrid - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria/Consejo Superior de Investigaciones Científicas (UPM-INIA/CSIC), Universidad Politécnica de Madrid, Madrid, Spain
| | - Araceli Diaz-Perales
- Centre for Plant Biotechnology and Genomics, Universidad Politécnica de Madrid - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria/Consejo Superior de Investigaciones Científicas (UPM-INIA/CSIC), Universidad Politécnica de Madrid, Madrid, Spain
- Department of Biotechnology-Plant Biology, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas (ETSIAAB), Universidad Politécnica de Madrid, Madrid, Spain
| | - Susan Waserman
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Manel Jordana
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Joshua F E Koenig
- Department of Medicine, Schroeder Allergy and Immunology Research Institute, McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
6
|
Li Y, Li W, Chen J, Qiu S, Liu Y, Xu L, Tian T, Li JP. Deciphering single-cell protein secretion and gene expressions by constructing cell-antibody conjugates. Bioorg Chem 2024; 143:106987. [PMID: 38039927 DOI: 10.1016/j.bioorg.2023.106987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/13/2023] [Accepted: 11/19/2023] [Indexed: 12/03/2023]
Abstract
Secreted proteins play critical roles in regulating immune responses, exerting cytotoxic effects on tumor cells, promoting inflammatory processes, and influencing cellular metabolism. Deciphering the intricate relationship between the heterogeneity of secreted proteins and their transcriptional states is pivotal in the study of cellular heterogeneity. Here we proposed a cell-antibody conjugate-based sequencing methodology (Cellab-seq) for joint characterization of secreted proteins and transcriptome. Cellab-seq utilizes a chemoenzymatic strategy to construct cell-antibody conjugates, which enables the capture of secreted proteins and their signal transduction with the incorporation of barcode detection antibodies. We applied Cellab-seq to investigate how gene expression influences the activity of secreted proteins in NK cells. Altogether, this strategy facilitates a nuanced understanding of cellular dynamics under diverse physiological conditions, ultimately contributing to the prevention, diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Yachao Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Wannan Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Jiashang Chen
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Shuang Qiu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yilong Liu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Lingjie Xu
- Vazyme Biotech, Red Maple Hi-tech Industry Park, Kechuang Road, Qixia District, Nanjing, Jiangsu 210023, China
| | - Tian Tian
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China.
| | - Jie P Li
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
7
|
Salimzadeh L, Burton AR, Le Bert N. Ex Vivo Fluorescent Labeling of HBV-Specific B Cells in Chronic Hepatitis B Patients. Methods Mol Biol 2024; 2837:241-255. [PMID: 39044090 DOI: 10.1007/978-1-0716-4027-2_21] [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: 07/25/2024]
Abstract
Fluorescently conjugated antigen-bait systems have been extensively used to identify antigen-specific B cells and probe humoral immunity across different settings. Following this approach, we used HBV antigens to bind the B cell receptor (BCR), permitting antigen-specific B cell detection by flow cytometry. Fluorochromes can either be attached covalently via chemical conjugation to the antigen or attached non-covalently by biotinylating the antigen. Dual-staining antigen-baits (where an antigen is directly conjugated to two distinct fluorochromes) have now been used to identify HBsAg- and HBcAg-specific B cells with a high degree of reliability and specificity. This system can be used to detect and characterize cells ex vivo or adapted to isolate antigen-specific cells using fluorescence-activated cell sorting.
Collapse
Affiliation(s)
- Loghman Salimzadeh
- Schwartz-Reisman Liver Research Centre, University Health Network, Toronto, ON, Canada.
- Toronto Centre for Liver Disease, University Health Network, Toronto, ON, Canada.
| | - Alice R Burton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK
| | - Nina Le Bert
- Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
8
|
Durgam SS, Khiew SH, Sayin I, Jain D, Yin D, Cavazzoni CB, Sage PT, King RG, Chong AS. MHC Tetramers Specifically Identify High- and Low-avidity Donor-specific B Cells in Transplantation Tolerance and Rejection. Transplantation 2023; 107:2526-2532. [PMID: 37493609 PMCID: PMC10811295 DOI: 10.1097/tp.0000000000004702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
BACKGROUND Although donor-specific antibody pre- and posttransplantation is routinely assessed, accurate quantification of memory alloreactive B cells that mediate recall antibody response remains challenging. Major histocompatibility complex (MHC) tetramers have been used to identify alloreactive B cells in mice and humans, but the specificity of this approach has not been rigorously assessed. METHODS B-cell receptors from MHC tetramer-binding single B cells were expressed as mouse recombinant immunoglobulin G1 (rIgG1) monoclonal antibodies, and the specificity was assessed with a multiplex bead assay. Relative binding avidity of rIgG1 was measured by modified dilution series technique and surface plasmon resonance. Additionally, immunoglobulin heavy chain variable regions of 50 individual B-cell receptors were sequenced to analyze the rate of somatic hypermutation. RESULTS The multiplex bead assay confirmed that expressed rIgG1 monoclonal antibodies were preferentially bound to bait MHC class II I-E d over control I-A d and I-A b tetramers. Furthermore, the dissociation constant 50 binding avidities of the rIgG1 ranged from 10 mM to 7 nM. The majority of tetramer-binding B cells were low avidity, and ~12.8% to 15.2% from naive and tolerant mice and 30.9% from acute rejecting mice were higher avidity (dissociation constant 50 <1 mM). CONCLUSIONS Collectively, these studies demonstrate that donor MHC tetramers, under stringent binding conditions with decoy self-MHC tetramers, can specifically identify a broad repertoire of donor-specific B cells under conditions of rejection and tolerance.
Collapse
Affiliation(s)
| | | | - Ismail Sayin
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| | - Dharmendra Jain
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| | - Dengping Yin
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| | - Cecilia B. Cavazzoni
- Transplantation Research Center, Renal Division, Brigham
and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham
and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - R. Glenn King
- Department of Microbiology, University of Alabama at
Birmingham, Birmingham, AL, 35294, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| |
Collapse
|
9
|
Akhtar M, Basher SR, Nizam NN, Hossain L, Bhuiyan TR, Qadri F, Lundgren A. T helper cell responses in adult diarrheal patients following natural infection with enterotoxigenic Escherichia coli are primarily of the Th17 type. Front Immunol 2023; 14:1220130. [PMID: 37809062 PMCID: PMC10552643 DOI: 10.3389/fimmu.2023.1220130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
Background Infection with enterotoxigenic Escherichia coli (ETEC) gives rise to IgA antibodies against both the heat labile toxin (LT) and colonization factors (CFs), which are considered to synergistically protect against ETEC diarrhea. Since the development of ETEC-specific long lived plasma cells and memory B cells is likely to be dependent on T helper (Th) cells, we investigated if natural ETEC diarrhea elicits ETEC-specific Th cells and their relation to IgA responses. Methods Th cell subsets were analyzed in adult Bangladeshi patients hospitalized due to ETEC diarrhea by flow cytometric analysis of peripheral blood mononuclear cells (PBMCs) isolated from blood collected day 2, 7, 30 and 90 after hospitalization as well as in healthy controls. The LT- and CF-specific Th responses were determined by analysis of IL-17A and IFN-γ in antigen stimulated PBMC cultures using ELISA. ETEC-specific IgA secreted by circulating antibody secreting cells (plasmablasts) were analyzed by using the antibodies in lymphocyte supernatants (ALS) ELISA-based method and plasma IgA was also measured by ELISA. Results ETEC patients mounted significant ALS and plasma IgA responses against LTB and CFs on day 7 after hospitalization. ETEC patients had significantly elevated proportions of memory Th cells with a Th17 phenotype (CCR6+CXCR3-) in blood compared to controls, while frequencies of Th1 (CCR6-CXCR3+) or Th2 (CCR6-CXCR3-) cells were not increased. Antigen stimulation of PBMCs revealed IL-17A responses to LT, most clearly observed after stimulation with double mutant heat labile toxin (dmLT), but also with LT B subunit (LTB), and to CS6 in samples from patients with LT+ or CS6+ ETEC bacteria. Some individuals also mounted IFN-γ responses to dmLT and LTB. Levels of LTB specific IgA antibodies in ALS, but not plasma samples correlated with both IL-17A (r=0.5, p=0.02) and IFN-γ (r=0.6, p=0.01) responses to dmLT. Conclusions Our results show that ETEC diarrhea induces T cell responses, which are predominantly of the Th17 type. The correlations between IL-17A and IFN-g and intestine-derived plasmablast responses support that Th responses may contribute to the development of protective IgA responses against ETEC infection. These observations provide important insights into T cell responses that need to be considered in the evaluation of advanced ETEC vaccine candidates.
Collapse
Affiliation(s)
- Marjahan Akhtar
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Salima Raiyan Basher
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Nuder Nower Nizam
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Lazina Hossain
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Divison, icddr, b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Anna Lundgren
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
10
|
Mbani CJ, Nekoua MP, Moukassa D, Hober D. The Fight against Poliovirus Is Not Over. Microorganisms 2023; 11:1323. [PMID: 37317297 DOI: 10.3390/microorganisms11051323] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/13/2023] [Accepted: 05/14/2023] [Indexed: 06/16/2023] Open
Abstract
Poliovirus (PV), the virus that causes both acute poliomyelitis and post-polio syndrome, is classified within the Enterovirus C species, and there are three wild PV serotypes: WPV1, WPV2 and WPV3. The launch of the Global Polio Eradication Initiative (GPEI) in 1988 eradicated two of the three serotypes of WPV (WPV2 and WPV3). However, the endemic transmission of WPV1 persists in Afghanistan and Pakistan in 2022. There are cases of paralytic polio due to the loss of viral attenuation in the oral poliovirus vaccine (OPV), known as vaccine-derived poliovirus (VDPV). Between January 2021 and May 2023, a total of 2141 circulating VDPV (cVDPV) cases were reported in 36 countries worldwide. Because of this risk, inactivated poliovirus (IPV) is being used more widely, and attenuated PV2 has been removed from OPV formulations to obtain bivalent OPV (containing only types 1 and 3). In order to avoid the reversion of attenuated OPV strains, the new OPV, which is more stable due to genome-wide modifications, as well as sabin IPV and virus-like particle (VLP) vaccines, is being developed and offers promising solutions for eradicating WP1 and VDPV.
Collapse
Affiliation(s)
- Chaldam Jespère Mbani
- Laboratoire de Virologie URL3610, Université de Lille, CHU Lille, 59000 Lille, France
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Technique, Université Marien Ngouabi, Brazzaville BP 69, Congo
| | | | - Donatien Moukassa
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Technique, Université Marien Ngouabi, Brazzaville BP 69, Congo
| | - Didier Hober
- Laboratoire de Virologie URL3610, Université de Lille, CHU Lille, 59000 Lille, France
| |
Collapse
|
11
|
Erdem R, De Coster I, Withanage K, Mercer LD, Marchant A, Taton M, Cools N, Lion E, Cassels F, Higgins D, Ivinson K, Locke E, Mahmood K, Wright PF, Gast C, White JA, Ackerman ME, Konopka-Anstadt JL, Mainou BA, Van Damme P. Safety, tolerability, and immunogenicity of inactivated poliovirus vaccine with or without E.coli double mutant heat-labile toxin (dmLT) adjuvant in healthy adults; a phase 1 randomized study. Vaccine 2023; 41:1657-1667. [PMID: 36746739 PMCID: PMC9996288 DOI: 10.1016/j.vaccine.2023.01.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/01/2023] [Accepted: 01/21/2023] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inactivated trivalent poliovirus vaccine (IPV) induces humoral immunity, which protects against paralytic poliomyelitis but does not induce sufficient mucosal immunity to block intestinal infection. We assessed the intestinal immunity in healthy adults in Belgium conferred by a co-formulation of IPV with the mucosal adjuvant double mutant Labile Toxin (dmLT) derived from Escherichia coli. METHODS Healthy fully IPV-vaccinated 18-45-year-olds were randomly allocated to three groups: on Day 1 two groups received one full dose of IPV (n = 30) or IPV + dmLT (n = 30) in a blinded manner, and the third received an open-label dose of bivalent live oral polio vaccine (bOPV types 1 and 3, n = 20). All groups received a challenge dose of bOPV on Day 29. Participants reported solicited and unsolicited adverse events (AE) using study diaries. Mucosal immune responses were measured by fecal neutralization and IgA on Days 29 and 43, with fecal shedding of challenge viruses measured for 28 days. Humoral responses were measured by serum neutralizing antibody (NAb). RESULTS Solicited and unsolicited AEs were mainly mild-to-moderate and transient in all groups, with no meaningful differences in rates between groups. Fecal shedding of challenge viruses in both IPV groups exceeded that of the bOPV group but was not different between IPV and IPV + dmLT groups. High serum NAb responses were observed in both IPV groups, alongside modest levels of fecal neutralization and IgA. CONCLUSIONS Addition of dmLT to IPV administered intramuscularly neither affected humoral nor intestinal immunity nor decreased fecal virus shedding following bOPV challenge. The tolerability of the dose of dmLT used in this study may allow higher doses to be investigated for impact on mucosal immunity. Registered on ClinicalTrials.gov - NCT04232943.
Collapse
Affiliation(s)
- Rahsan Erdem
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Ilse De Coster
- Vaccine & Infectious Disease Institute, Centre for the Evaluation of Vaccination, University of Antwerp, Edegem, Belgium.
| | - Kanchanamala Withanage
- Vaccine & Infectious Disease Institute, Centre for the Evaluation of Vaccination, University of Antwerp, Edegem, Belgium
| | - Laina D Mercer
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Brussels, Belgium
| | - Martin Taton
- Institute for Medical Immunology, Université libre de Bruxelles, Brussels, Belgium
| | - Nathalie Cools
- Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
| | - Eva Lion
- Vaccine & Infectious Disease Institute, Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Belgium
| | - Fred Cassels
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Deborah Higgins
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Karen Ivinson
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Emily Locke
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Kutub Mahmood
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | | | - Chris Gast
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | - Jessica A White
- PATH, Center for Vaccine Innovation and Access, Seattle, WA, USA
| | | | | | - Bernardo A Mainou
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Pierre Van Damme
- Vaccine & Infectious Disease Institute, Centre for the Evaluation of Vaccination, University of Antwerp, Edegem, Belgium
| |
Collapse
|
12
|
Tsai CJY, Loh JMS, Fujihashi K, Kiyono H. Mucosal vaccination: onward and upward. Expert Rev Vaccines 2023; 22:885-899. [PMID: 37817433 DOI: 10.1080/14760584.2023.2268724] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION The unique mucosal immune system allows the generation of robust protective immune responses at the front line of pathogen encounters. The needle-free delivery route and cold chain-free logistic requirements also provide additional advantages in ease and economy. However, the development of mucosal vaccines faces several challenges, and only a handful of mucosal vaccines are currently licensed. These vaccines are all in the form of live attenuated or inactivated whole organisms, whereas no subunit-based mucosal vaccine is available. AREAS COVERED The selection of antigen, delivery vehicle, route and adjuvants for mucosal vaccination are highly important. This is particularly crucial for subunit vaccines, as they often fail to elicit strong immune responses. Emerging research is providing new insights into the biological and immunological uniqueness of mucosal tissues. However, many aspects of the mucosal immunology still await to be investigated. EXPERT OPINION This article provides an overview of the current understanding of mucosal vaccination and discusses the remaining knowledge gaps. We emphasize that because of the potential benefits mucosal vaccines can bring from the biomedical, social and economic standpoints, the unmet goal to achieve mucosal vaccine success is worth the effort.
Collapse
Affiliation(s)
- Catherine J Y Tsai
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan
| |
Collapse
|
13
|
Palacios-Pedrero MÁ, Jansen JM, Blume C, Stanislawski N, Jonczyk R, Molle A, Hernandez MG, Kaiser FK, Jung K, Osterhaus ADME, Rimmelzwaan GF, Saletti G. Signs of immunosenescence correlate with poor outcome of mRNA COVID-19 vaccination in older adults. NATURE AGING 2022; 2:896-905. [PMID: 37118289 PMCID: PMC10154205 DOI: 10.1038/s43587-022-00292-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/02/2022] [Indexed: 04/30/2023]
Abstract
Vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is effective in preventing COVID-19 hospitalization and fatal outcome. However, several studies indicated that there is reduced vaccine effectiveness among older individuals, which is correlated with their general health status1,2. How and to what extent age-related immunological defects are responsible for the suboptimal vaccine responses observed in older individuals receiving SARS-CoV-2 messenger RNA vaccine, is unclear and not fully investigated1,3-5. In this observational study, we investigated adaptive immune responses in adults of various ages (22-99 years old) receiving 2 doses of the BNT162b2 mRNA vaccine. Vaccine-induced Spike-specific antibody, and T and memory B cell responses decreased with increasing age. These responses positively correlated with the percentages of peripheral naïve CD4+ and CD8+ T cells and negatively with CD8+ T cells expressing signs of immunosenescence. Older adults displayed a preferred T cell response to the S2 region of the Spike protein, which is relatively conserved and a target for cross-reactive T cells induced by human 'common cold' coronaviruses. Memory T cell responses to influenza virus were not affected by age-related changes, nor the SARS-CoV-2-specific response induced by infection. Collectively, we identified signs of immunosenescence correlating with the outcome of vaccination against a new viral antigen to which older adults are immunologically naïve. This knowledge is important for the management of COVID-19 infections in older adults.
Collapse
Affiliation(s)
| | - Janina M Jansen
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Cornelia Blume
- Institute of Technical Chemistry, Leibniz University, Hanover, Germany
| | - Nils Stanislawski
- Institute of Microelectronic Systems, Leibniz University, Hanover, Germany
| | - Rebecca Jonczyk
- Institute of Technical Chemistry, Leibniz University, Hanover, Germany
| | - Antonia Molle
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Mariana Gonzalez Hernandez
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Franziska K Kaiser
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, Genomics and Bioinformatics of Infectious Diseases, University of Veterinary Medicine, Hanover, Germany
| | - Albert D M E Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany
- Global Virus Network, Center of Excellence, Buffalo, NY, USA
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany.
| | - Giulietta Saletti
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hanover, Germany.
| |
Collapse
|
14
|
Seth A, Mittal E, Luan J, Kolla S, Mazer MB, Joshi H, Gupta R, Rathi P, Wang Z, Morrissey JJ, Ernst JD, Portal-Celhay C, Morley SC, Philips JA, Singamaneni S. High-resolution imaging of protein secretion at the single-cell level using plasmon-enhanced FluoroDOT assay. CELL REPORTS METHODS 2022; 2:100267. [PMID: 36046626 PMCID: PMC9421537 DOI: 10.1016/j.crmeth.2022.100267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/28/2022] [Accepted: 07/13/2022] [Indexed: 11/30/2022]
Abstract
Secreted proteins mediate essential physiological processes. With conventional assays, it is challenging to map the spatial distribution of proteins secreted by single cells, to study cell-to-cell heterogeneity in secretion, or to detect proteins of low abundance or incipient secretion. Here, we introduce the "FluoroDOT assay," which uses an ultrabright nanoparticle plasmonic-fluor that enables high-resolution imaging of protein secretion. We find that plasmonic-fluors are 16,000-fold brighter, with nearly 30-fold higher signal-to-noise compared with conventional fluorescence labels. We demonstrate high-resolution imaging of different secreted cytokines in the single-plexed and spectrally multiplexed FluoroDOT assay that revealed cellular heterogeneity in secretion of multiple proteins simultaneously. Using diverse biochemical stimuli, including Mycobacterium tuberculosis infection, and a variety of immune cells such as macrophages, dendritic cells (DCs), and DC-T cell co-culture, we demonstrate that the assay is versatile, facile, and widely adaptable for enhancing biological understanding of spatial and temporal dynamics of single-cell secretome.
Collapse
Affiliation(s)
- Anushree Seth
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Auragent Bioscience, LLC, St. Louis, MO 63108, USA
| | - Ekansh Mittal
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63130, USA
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Jingyi Luan
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Samhitha Kolla
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Monty B. Mazer
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hemant Joshi
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rohit Gupta
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Priya Rathi
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Zheyu Wang
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Jeremiah J. Morrissey
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Joel D. Ernst
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Cynthia Portal-Celhay
- Division of Infectious Diseases, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Sharon Celeste Morley
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer A. Philips
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63130, USA
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
15
|
Divergent age-related humoral correlates of protection against respiratory syncytial virus infection in older and young adults: a pilot, controlled, human infection challenge model. THE LANCET. HEALTHY LONGEVITY 2022; 3:e405-e416. [PMID: 36098319 DOI: 10.1016/s2666-7568(22)00103-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Respiratory viral infections are typically more severe in older adults. Older adults are more vulnerable to infection and do not respond effectively to vaccines due to a combination of immunosenescence, so-called inflamm-ageing, and accumulation of comorbidities. Although age-related changes in immune responses have been described, the causes of this enhanced respiratory disease in older adults remain poorly understood. We therefore performed volunteer challenge with respiratory syncytial virus (RSV) in groups of younger and older adult volunteers. The aim of this study was to establish the safety and tolerability of this model and define age-related clinical, virological, and immunological outcomes. METHODS In this human infection challenge pilot study, adults aged 18-55 years and 60-75 years were assessed for enrolment using protocol-defined inclusion and exclusion criteria. Symptoms were documented by self-completed diaries and viral load determined by quantitative PCR of nasal lavage. Peripheral blood B cell frequencies were measured by enzyme-linked immunospot and antibodies against pre-fusion and post-fusion, NP, and G proteins in the blood and upper respiratory tract were measured. The study was registered with ClinicalTrials.gov, NCT03728413. FINDINGS 381 adults aged 60-75 years (older cohort) and 19 adults aged 18-55 years (young cohort) were assessed for enrolment using protocol-defined inclusion and exclusion criteria between Nov 12, 2018, and Feb 26, 2020. 12 healthy volunteers aged 60-75 years and 21 aged 18-55 years were inoculated intranasally with RSV Memphis-37. Nine (67%) of the 12 older volunteers became infected, developing mild-to-moderate upper respiratory tract symptoms that resolved without serious adverse events or sequelae. Viral load peaked on day 6 post-inoculation and symptoms peaked between days 6 and 8. Increases in circulating IgG-positive and IgA-positive antigen-specific plasmablasts, serum neutralising antibodies, and pre-F specific IgG were similar younger and older adults. However, in contrast to young participants, secretory IgA titres in older volunteers failed to increase during infection and, unlike serum IgG, did not correlate with protection. INTERPRETATION Better understanding of age-related differences in clinical outcomes and immune correlates of protection can overcome reduction in vaccine efficacy with advancing age. We identify correlates of protection in older adults, revealing previously unrecognised factors which might have implications for targeted vaccine discovery and drug development in this vulnerable group. FUNDING Medical Research Council and GlaxoSmithKline EMINENT Consortium.
Collapse
|
16
|
MARTINUZZI E, BENZAQUEN J, GUERIN O, LEROY S, SIMON T, ILIE M, HOFMAN V, ALLEGRA M, TANGA V, MICHEL E, BOUTROS J, MANIEL C, SICARD A, GLAICHENHAUS N, CZERKINSKY C, BLANCOU P, HOFMAN P, MARQUETTE CH. A Single Dose of BNT162b2 Messenger RNA Vaccine Induces Airway Immunity in Severe Acute Respiratory Syndrome Coronavirus 2 Naive and Recovered Coronavirus Disease 2019 Subjects. Clin Infect Dis 2022; 75:2053-2059. [PMID: 35579991 PMCID: PMC9129216 DOI: 10.1093/cid/ciac378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Mucosal antibodies can prevent virus entry and replication in mucosal epithelial cells and therefore virus shedding. Parenteral booster injection of a vaccine against a mucosal pathogen promotes stronger mucosal immune responses following prior mucosal infection compared with injections of a parenteral vaccine in a mucosally naive subject. We investigated whether this was also the case for the BNT162b2 coronavirus disease 2019 (COVID-19) messenger RNA vaccine. METHODS Twenty recovered COVID-19 subjects (RCSs) and 23 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-naive subjects were vaccinated with, respectively, 1 and 2 doses of the BNT162b2 COVID-19 vaccine. Nasal epithelial lining fluid (NELF) and plasma were collected before and after vaccination and assessed for immunoglobulin G (IgG) and IgA antibody levels to Spike and for their ability to neutralize binding of Spike to angiotensin-converting enzyme-2 receptor. Blood was analyzed 1 week after vaccination for the number of Spike-specific antibody-secreting cells (ASCs) with a mucosal tropism. RESULTS All RCSs had both nasal and blood SARS-CoV-2-specific antibodies at least 90 days after initial diagnosis. In RCSs, a single dose of vaccine amplified preexisting Spike-specific IgG and IgA antibody responses in both NELF and blood against both vaccine homologous and variant strains, including Delta. These responses were associated with Spike-specific IgG and IgA ASCs with a mucosal tropism in blood. Nasal IgA and IgG antibody responses were lower in magnitude in SARS-CoV-2-naive subjects after 2 vaccine doses compared with RCSs after 1 dose. CONCLUSIONS Mucosal immune response to the SARS-CoV-2 Spike protein is higher in RCSs after a single vaccine dose compared with SARS-CoV-2-naive subjects after 2 doses.
Collapse
Affiliation(s)
- Emanuela MARTINUZZI
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Jonathan BENZAQUEN
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, Nice, France,Université Côte d'Azur, CNRS, INSERM, Institute of Research on Cancer and Aging, Nice, France
| | - Olivier GUERIN
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Pôle Réhabilitation Autonomie Vieillissement, Nice, France
| | - Sylvie LEROY
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, Nice, France
| | - Thomas SIMON
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Marius ILIE
- Université Côte d'Azur, CNRS, INSERM, Institute of Research on Cancer and Aging, Nice, France,Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Laboratory of Clinical and Experimental Pathology, Biobank (BB-0033-00025), FHU OncoAge, Centre Nice, France
| | - Véronique HOFMAN
- Université Côte d'Azur, CNRS, INSERM, Institute of Research on Cancer and Aging, Nice, France,Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Laboratory of Clinical and Experimental Pathology, Biobank (BB-0033-00025), FHU OncoAge, Centre Nice, France
| | - Maryline ALLEGRA
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Laboratory of Clinical and Experimental Pathology, Biobank (BB-0033-00025), FHU OncoAge, Centre Nice, France
| | - Virginie TANGA
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Laboratory of Clinical and Experimental Pathology, Biobank (BB-0033-00025), FHU OncoAge, Centre Nice, France
| | - Emeline MICHEL
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Pôle Réhabilitation Autonomie Vieillissement, Nice, France
| | - Jacques BOUTROS
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, Nice, France
| | - Charlotte MANIEL
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, Nice, France
| | - Antoine SICARD
- University Côte d’Azur, Clinical Research Unit Côte d’Azur, Nice, France
| | - Nicolas GLAICHENHAUS
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France,University Côte d’Azur, Clinical Research Unit Côte d’Azur, Nice, France
| | - Cecil CZERKINSKY
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France,Alternate corresponding author in the event that the corresponding author is unavailable: Cecil Czerkinsky, Md, PhD, Nice, France ()
| | - Philippe BLANCOU
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Paul HOFMAN
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France,Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Laboratory of Clinical and Experimental Pathology, Biobank (BB-0033-00025), FHU OncoAge, Centre Nice, France
| | - Charles H. MARQUETTE
- Université Côte d'Azur, Centre Hospitalier Universitaire de Nice, Department of Pulmonary Medicine and Thoracic Oncology, FHU OncoAge, Nice, France,Université Côte d'Azur, CNRS, INSERM, Institute of Research on Cancer and Aging, Nice, France,Corresponding author: Charles H Marquette, Md, PhD, Nice, France ()
| |
Collapse
|
17
|
Broketa M, Bruhns P. Single-Cell Technologies for the Study of Antibody-Secreting Cells. Front Immunol 2022; 12:821729. [PMID: 35173713 PMCID: PMC8841722 DOI: 10.3389/fimmu.2021.821729] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/29/2021] [Indexed: 01/05/2023] Open
Abstract
Antibody-secreting cells (ASC), plasmablasts and plasma cells, are terminally differentiated B cells responsible for large-scale production and secretion of antibodies. ASC are derived from activated B cells, which may differentiate extrafollicularly or form germinal center (GC) reactions within secondary lymphoid organs. ASC therefore consist of short-lived, poorly matured plasmablasts that generally secrete lower-affinity antibodies, or long-lived, highly matured plasma cells that generally secrete higher-affinity antibodies. The ASC population is responsible for producing an immediate humoral B cell response, the polyclonal antibody repertoire, as well as in parallel building effective humoral memory and immunity, or potentially driving pathology in the case of autoimmunity. ASC are phenotypically and transcriptionally distinct from other B cells and further distinguishable by morphology, varied lifespans, and anatomical localization. Single cell analyses are required to interrogate the functional and transcriptional diversity of ASC and their secreted antibody repertoire and understand the contribution of individual ASC responses to the polyclonal humoral response. Here we summarize the current and emerging functional and molecular techniques for high-throughput characterization of ASC with single cell resolution, including flow and mass cytometry, spot-based and microfluidic-based assays, focusing on functional approaches of the secreted antibodies: specificity, affinity, and secretion rate.
Collapse
Affiliation(s)
- Matteo Broketa
- Institut Pasteur, Université de Paris, INSERM UMR 1222, Unit of Antibodies in Therapy and Pathology, Paris, France
- Sorbonne Université, Collège doctoral, Paris, France
| | - Pierre Bruhns
- Institut Pasteur, Université de Paris, INSERM UMR 1222, Unit of Antibodies in Therapy and Pathology, Paris, France
| |
Collapse
|
18
|
Venkatesan MM, Ballou C, Barnoy S, McNeal M, El-Khorazaty J, Frenck R, Baqar S. Antibody in Lymphocyte Supernatant (ALS) responses after oral vaccination with live Shigella sonnei vaccine candidates WRSs2 and WRSs3 and correlation with serum antibodies, ASCs, fecal IgA and shedding. PLoS One 2021; 16:e0259361. [PMID: 34793505 PMCID: PMC8601580 DOI: 10.1371/journal.pone.0259361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/18/2021] [Indexed: 12/15/2022] Open
Abstract
The levels of antigen-specific Antibodies in Lymphocyte Supernatant (ALS) using an ELISA are being used to evaluate mucosal immune responses as an alternate to measuring the number of Antibody Secreting Cells (ASCs) using an ELISpot assay. A recently completed trial of two novel S. sonnei live oral vaccine candidates WRSs2 and WRSs3 established that both candidates were safe, well tolerated and immunogenic in a vaccine dose-dependent manner. Previously, mucosal immune responses were measured by assaying IgA- and IgG-ASC in peripheral blood mononuclear cells (PBMCs). In this report, the magnitude of the S. sonnei antigen-specific IgA- and IgG-ALS responses was measured and correlated with previously described ASCs, serum antibodies, fecal IgA and vaccine shedding. Overall, the magnitude of S. sonnei anti-Invaplex50 ALS was higher than that of LPS or IpaB, and both vaccines demonstrated a more robust IgA-ALS response than IgG; however, compared to WRSs3, the magnitude and percentage of responders were higher among WRSs2 recipients for IgA- or IgG-ALS. All WRSs2 vaccinees at the two highest doses responded for LPS and Invaplex50-specific IgA-ALS and 63-100% for WRSs3 vaccinees responded. Regardless of the vaccine candidate, vaccine dose or detecting antigen, the kinetics of ALS responses were similar peaking on days 7 to 9 and returning to baseline by day 14. The ALS responses were vaccine-specific since no responses were detected among placebo recipients at any time. A strong correlation and agreement between responders/non-responders were noted between ALS and other mucosal (ASC and fecal IgA) and systemic (serum antibody) immune responses. These data indicate that the ALS assay can be a useful tool to evaluate mucosal responses to oral vaccination, an observation noted with trials of other bacterial diarrheal pathogens. Furthermore, this data will guide the list of immunological assays to be conducted for efficacy trials in different populations. It is hoped that an antigen-specific-ALS titer may be a key mucosal correlate of protection, a feature not currently available for any Shigella vaccines candidates. https://clinicaltrials.gov/show/NCT01336699.
Collapse
Affiliation(s)
- Malabi M. Venkatesan
- Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | | | - Shoshana Barnoy
- Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Monica McNeal
- Division of Infectious Diseases, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | | | - Robert Frenck
- Division of Infectious Diseases, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Shahida Baqar
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
19
|
Immunological Assessment of Lung Responses to Inhalational Lipoprotein Vaccines Against Bacterial Pathogens. Methods Mol Biol 2021. [PMID: 34784043 DOI: 10.1007/978-1-0716-1900-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Lipopeptides or lipoproteins show potential as safe and effective subunit vaccines for protection against bacterial pathogens. Provided suitable adjuvants are selected, such as the TLR2-stimulating molecules Pam2Cys and Pam3Cys, these may be formulated as inhalational vaccines to optimize localized pulmonary immune responses. Here, we present methods to assess antigen-specific memory lymphocyte responses to novel vaccines, with a focus on immune responses in the lung tissue and bronchoalveolar space. We describe detection of T-cell responses via leukocyte restimulation, followed by intracellular cytokine staining and flow cytometry, enzyme-linked immunosorbent spot assay (ELISpot), and sustained leukocyte restimulation for detection of antigen-specific memory responses. We also detail assessment of antibody responses to vaccine antigens, via enzyme-linked immunosorbent assay (ELISA)-based detection. These methods are suitable for testing a wide range of pulmonary vaccines.
Collapse
|
20
|
Hantavirus infection-induced B cell activation elevates free light chains levels in circulation. PLoS Pathog 2021; 17:e1009843. [PMID: 34379707 PMCID: PMC8382192 DOI: 10.1371/journal.ppat.1009843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/23/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
In humans, orthohantaviruses can cause hemorrhagic fever with renal syndrome (HFRS) or hantavirus pulmonary syndrome (HPS). An earlier study reported that acute Andes virus HPS caused a massive and transient elevation in the number of circulating plasmablasts with specificity towards both viral and host antigens suggestive of polyclonal B cell activation. Immunoglobulins (Igs), produced by different B cell populations, comprise heavy and light chains; however, a certain amount of free light chains (FLCs) is constantly present in serum. Upregulation of FLCs, especially clonal species, associates with renal pathogenesis by fibril or deposit formations affecting the glomeruli, induction of epithelial cell disorders, or cast formation in the tubular network. We report that acute orthohantavirus infection increases the level of Ig FLCs in serum of both HFRS and HPS patients, and that the increase correlates with the severity of acute kidney injury in HFRS. The fact that the kappa to lambda FLC ratio in the sera of HFRS and HPS patients remained within the normal range suggests polyclonal B cell activation rather than proliferation of a single B cell clone. HFRS patients demonstrated increased urinary excretion of FLCs, and we found plasma cell infiltration in archival patient kidney biopsies that we speculate to contribute to the observed FLC excreta. Analysis of hospitalized HFRS patients’ peripheral blood mononuclear cells showed elevated plasmablast levels, a fraction of which stained positive for Puumala virus antigen. Furthermore, B cells isolated from healthy donors were susceptible to Puumala virus in vitro, and the virus infection induced increased production of Igs and FLCs. The findings propose that hantaviruses directly activate B cells, and that the ensuing intense production of polyclonal Igs and FLCs may contribute to acute hantavirus infection-associated pathological findings. Orthohantaviruses are globally spread zoonotic pathogens, which can cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS) with significant burden to human health. The pathogenesis mechanisms of orthohantavirus-caused diseases are not known in detail; however, excessive immune response towards the virus with concomitant pathological effects against host tissues appears to be a contributing factor. Here we report an increase of free immunoglobulin (Ig) light chains (FLCs), components required to make complete Ig molecules, in blood of acute HFRS and HPS. Samples collected during acute HFRS demonstrated increased FLCs levels in the urine and blood of patients hospitalized due the disease. Furthermore, the FLC levels positively correlated with markers of acute kidney injury. In addition, our results show that orthohantaviruses can infect and activate B cells to produce FLCs as well as whole Igs, which provides a mechanistic explanation of the increased FLC levels in patients. Taken together, our results suggest that aberrant antibody responses might play a role in the pathogenesis of orthohantavirus infections.
Collapse
|
21
|
Abstract
Mucosal vaccines offer the potential to trigger robust protective immune responses at the predominant sites of pathogen infection. In principle, the induction of adaptive immunity at mucosal sites, involving secretory antibody responses and tissue-resident T cells, has the capacity to prevent an infection from becoming established in the first place, rather than only curtailing infection and protecting against the development of disease symptoms. Although numerous effective mucosal vaccines are in use, the major advances seen with injectable vaccines (including adjuvanted subunit antigens, RNA and DNA vaccines) have not yet been translated into licensed mucosal vaccines, which currently comprise solely live attenuated and inactivated whole-cell preparations. The identification of safe and effective mucosal adjuvants allied to innovative antigen discovery and delivery strategies is key to advancing mucosal vaccines. Significant progress has been made in resolving the mechanisms that regulate innate and adaptive mucosal immunity and in understanding the crosstalk between mucosal sites, and this provides valuable pointers to inform mucosal adjuvant design. In particular, increased knowledge on mucosal antigen-presenting cells, innate lymphoid cell populations and resident memory cells at mucosal sites highlights attractive targets for vaccine design. Exploiting these insights will allow new vaccine technologies to be leveraged to facilitate rational mucosal vaccine design for pathogens including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and for cancer.
Collapse
|
22
|
Stensland ZC, Smith MJ. Enrichment and Detection of Antigen-Binding B Cells for Mass Cytometry. MAGNETOCHEMISTRY (BASEL, SWITZERLAND) 2021; 7:92. [PMID: 34295938 PMCID: PMC8294334 DOI: 10.3390/magnetochemistry7070092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Over the years, various techniques have been utilized to study the function and phenotype of antigen-binding B cells in the primary repertoire following immunization, infection, and development of autoimmunity. Due to the low frequency of antigen-reactive B cells (<0.05% of lymphocytes) in the periphery, preliminary enrichment of cells is necessary to achieve sufficient numbers for statistically sound characterization, especially when downstream analytic platform use, e.g., CyTOF, is low throughput. We previously described a method to detect and enrich antigen-reactive B cells from peripheral blood and tissues using biotinylated antigens in conjunction with magnetic nanoparticles, preparative to a downstream analysis by ELISPOT and flow cytometry. While mass cytometry (CyTOF) enables high dimensional immunophenotyping of over 40 unique parameters on a single-cell level, its low throughput compared to flow cytometry and requirement for removal of metal contaminants, such as nanoparticles, made it particularly unsuitable for studies of rare cells in a mixed population. Here we describe a novel CyTOF-compatible approach for multiplexed enrichment of antigen-reactive B cells, e.g., insulin and tetanus toxoid, using cleavable magnetic nanoparticles. This method allows improved monitoring of the phenotype and function of antigen-reactive B cells during the development of disease or after immunization while minimizing the amount of sample and run times needed.
Collapse
Affiliation(s)
- Zachary C. Stensland
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mia J. Smith
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
23
|
Ticha O, Klemm D, Moos L, Bekeredjian-Ding I. A cell-based in vitro assay for testing of immunological integrity of Tetanus toxoid vaccine antigen. NPJ Vaccines 2021; 6:88. [PMID: 34162886 PMCID: PMC8222253 DOI: 10.1038/s41541-021-00344-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 05/24/2021] [Indexed: 12/03/2022] Open
Abstract
Vaccines containing inactivated toxins confer protection by eliciting a neutralizing antibody response against bacterial toxins such as tetanus and diphtheria. At present, release of tetanus toxoid (TT) and diphtheria toxoid (DT)-containing vaccines relies on in vivo experiments showing the protective vaccine response. The aim of this study was to develop a reliable in vitro assay for TT vaccine antigen characterization with the potential of replacing in vivo potency experiments. To this end, we exploited that TT elicits a recall response in vaccinated donors: human peripheral blood mononuclear cells (PBMC) were stimulated with alum-adsorbed TT bulk antigen and low concentrations of TLR9 ligand; induction of TT-specific IgG was quantified via ELISpot after 5 days. Proof-of-concept was obtained using paired samples from donors before and after vaccination; anti-TT IgG was only detected in PBMC collected after booster vaccination; specificity was demonstrated with DT stimulation as control. Notably, when using PBMC from buffy coats, the specific response to TT was reproducible in 30% of cells; responsiveness correlated with higher numbers of switched memory B cells. Consecutive results showed that TT-specific IgG was also detectable when PBMC were stimulated with DTaP final vaccine product. Thus, the assay provides a viable means to test B-cell differentiation and induction of TT-specific IgG secretion using bulk antigen and final vaccine. However, prequalification of PBMC is required for reliable performance. Along with physicochemical and immunochemical methods, the functional assay could represent a complementary tool to replace in vivo potency assays in batch release of TT-containing vaccines.
Collapse
Affiliation(s)
- Olga Ticha
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | - Dido Klemm
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | - Lukas Moos
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | | |
Collapse
|
24
|
Zhou Y, Shao N, Bessa de Castro R, Zhang P, Ma Y, Liu X, Huang F, Wang RF, Qin L. Evaluation of Single-Cell Cytokine Secretion and Cell-Cell Interactions with a Hierarchical Loading Microwell Chip. Cell Rep 2021; 31:107574. [PMID: 32348757 PMCID: PMC7583657 DOI: 10.1016/j.celrep.2020.107574] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/21/2020] [Accepted: 04/02/2020] [Indexed: 02/01/2023] Open
Abstract
Comprehensive evaluation of single T cell functions such as cytokine secretion and cytolysis of target cells is greatly needed in adoptive cell therapy (ACT) but has never been fully fulfilled by current approaches. Herein, we develop a hierarchical loading microwell chip (HL-Chip) that aligns multiple cells and functionalized beads in a high-throughput microwell array with single-cell/bead precision based on size differences. We demonstrate the potential of the HL-Chip in evaluating single T cell functions by three applications: high-throughput longitudinal secretory profiling of single T cells, large-scale evaluation of cytolytic activity of single T cells, and integrated T cell-tumor cell interactions. The HL-Chip is a simple and robust technology that constructs arrays of defined cell/object combinations for multiple measurements and material retrieval.
Collapse
Affiliation(s)
- Yufu Zhou
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; The Third Xiangya Hospital, Central South University, Changsha 410008, China; Center for inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Ning Shao
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ricardo Bessa de Castro
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Pengchao Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Yuan Ma
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xin Liu
- Center for inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine and Norris Comprehensive Cancer Center, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Feizhou Huang
- The Third Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rong-Fu Wang
- Center for inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Pediatrics, Children's Hospital of Los Angeles, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA; Department of Medicine and Norris Comprehensive Cancer Center, The Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
25
|
Bucheli OTM, Sigvaldadóttir I, Eyer K. Measuring single-cell protein secretion in immunology: Technologies, advances, and applications. Eur J Immunol 2021; 51:1334-1347. [PMID: 33734428 PMCID: PMC8252417 DOI: 10.1002/eji.202048976] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/12/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
The dynamics, nature, strength, and ultimately protective capabilities of an active immune response are determined by the extracellular constitution and concentration of various soluble factors. Generated effector cells secrete such mediators, including antibodies, chemo‐ and cytokines to achieve functionality. These secreted factors organize the individual immune cells into functional tissues, initiate, orchestrate, and regulate the immune response. Therefore, a single‐cell resolved analysis of protein secretion is a valuable tool for studying the heterogeneity and functionality of immune cells. This review aims to provide a comparative overview of various methods to characterize immune reactions by measuring single‐cell protein secretion. Spot‐based and cytometry‐based assays, such as ELISpot and flow cytometry, respectively, are well‐established methods applied in basic research and clinical settings. Emerging novel technologies, such as microfluidic platforms, offer new ways to measure and exploit protein secretion in immune reactions. Further technological advances will allow the deciphering of protein secretion in immunological responses with unprecedented detail, linking secretion to functionality. Here, we summarize the development and recent advances of tools that allow the analysis of protein secretion at the single‐cell level, and discuss and contrast their applications within immunology.
Collapse
Affiliation(s)
- Olivia T M Bucheli
- ETH Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland
| | - Ingibjörg Sigvaldadóttir
- ETH Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland
| | - Klaus Eyer
- ETH Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
26
|
Mottram L, Lundgren A, Svennerholm AM, Leach S. A Systems Biology Approach Identifies B Cell Maturation Antigen (BCMA) as a Biomarker Reflecting Oral Vaccine Induced IgA Antibody Responses in Humans. Front Immunol 2021; 12:647873. [PMID: 33828557 PMCID: PMC8019727 DOI: 10.3389/fimmu.2021.647873] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Vaccines against enteric diseases could improve global health. Despite this, only a few oral vaccines are currently available for human use. One way to facilitate such vaccine development could be to identify a practical and relatively low cost biomarker assay to assess oral vaccine induced primary and memory IgA immune responses in humans. Such an IgA biomarker assay could complement antigen-specific immune response measurements, enabling more oral vaccine candidates to be tested, whilst also reducing the work and costs associated with early oral vaccine development. With this in mind, we take a holistic systems biology approach to compare the transcriptional signatures of peripheral blood mononuclear cells isolated from volunteers, who following two oral priming doses with the oral cholera vaccine Dukoral®, had either strong or no vaccine specific IgA responses. Using this bioinformatical method, we identify TNFRSF17, a gene encoding the B cell maturation antigen (BCMA), as a candidate biomarker of oral vaccine induced IgA immune responses. We then assess the ability of BCMA to reflect oral vaccine induced primary and memory IgA responses using an ELISA BCMA assay on a larger number of samples collected in clinical trials with Dukoral® and the oral enterotoxigenic Escherichia coli vaccine candidate ETVAX. We find significant correlations between levels of BCMA and vaccine antigen-specific IgA in antibodies in lymphocyte secretion (ALS) specimens, as well as with proportions of circulating plasmablasts detected by flow cytometry. Importantly, our results suggest that levels of BCMA detected early after primary mucosal vaccination may be a biomarker for induction of long-lived vaccine specific memory B cell responses, which are otherwise difficult to measure in clinical vaccine trials. In addition, we find that ALS-BCMA responses in individuals vaccinated with ETVAX plus the adjuvant double mutant heat-labile toxin (dmLT) are significantly higher than in subjects given ETVAX only. We therefore propose that as ALS-BCMA responses may reflect the total vaccine induced IgA responses to oral vaccination, this BCMA ELISA assay could also be used to estimate the total adjuvant effect on vaccine induced-antibody responses, independently of antigen specificity, further supporting the usefulness of the assay.
Collapse
Affiliation(s)
- Lynda Mottram
- Gothenburg University Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Lundgren
- Gothenburg University Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ann-Mari Svennerholm
- Gothenburg University Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susannah Leach
- Gothenburg University Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Pharmacology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
27
|
Meyer Sauteur PM, Trück J, van Rossum AMC, Berger C. Circulating Antibody-Secreting Cell Response During Mycoplasma pneumoniae Childhood Pneumonia. J Infect Dis 2021; 222:136-147. [PMID: 32034406 DOI: 10.1093/infdis/jiaa062] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/06/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND We recently demonstrated that the measurement of Mycoplasma pneumoniae (Mp)-specific immunoglobulin (Ig)M antibody-secreting cells (ASCs) improved diagnosis of Mp infection. Here, we aimed to describe Mp ASC kinetics and duration in comparison to conventional measures such as pharyngeal Mp deoxyribonucleic acid (DNA) and serum antibodies. METHODS This is a prospective longitudinal study of 63 community-acquired pneumonia (CAP) patients and 21 healthy controls (HCs), 3-18 years of age, from 2016 to 2017. Mycoplasma pneumoniae ASCs measured by enzyme-linked immunospot assay were assessed alongside Mp DNA and antibodies during 6-month follow-up. RESULTS Mycoplasma pneumoniae ASCs of the isotype IgM were found in 29 (46%), IgG were found in 27 (43%), and IgA were found in 27 (43%) CAP patients. Mycoplasma pneumoniae ASCs were detected from 2 days to a maximum of 6 weeks after symptom onset, whereas Mp DNA and antibodies persisted until 4 months (P = .03) and 6 months (P < .01). Mycoplasma pneumoniae ASCs were undetectable in HCs, in contrast to detection of Mp DNA in 10 (48%) or antibodies in 6 (29%) controls for a prolonged time. The Mp ASC response correlated with clinical disease, but it did not differ between patients treated with or without antibiotics against Mp. CONCLUSIONS Mycoplasma pneumoniae-specific ASCs are short-lived and associated with clinical disease, making it an optimal resource for determining Mp pneumonia etiology.
Collapse
Affiliation(s)
- Patrick M Meyer Sauteur
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Johannes Trück
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland.,Division of Immunology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Annemarie M C van Rossum
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, Erasmus MC University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Christoph Berger
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Heo M, Chenon G, Castrillon C, Bibette J, Bruhns P, Griffiths AD, Baudry J, Eyer K. Deep phenotypic characterization of immunization-induced antibacterial IgG repertoires in mice using a single-antibody bioassay. Commun Biol 2020; 3:614. [PMID: 33106526 PMCID: PMC7589517 DOI: 10.1038/s42003-020-01296-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Antibodies with antibacterial activity need to bind to the bacterial surface with affinity, specificity, and sufficient density to induce efficient elimination. To characterize the anti-bacterial antibody repertoire, we developed an in-droplet bioassay with single-antibody resolution. The assay not only allowed us to identify whether the secreted antibodies recognized a bacterial surface antigen, but also to estimate the apparent dissociation constant (KD app) of the interaction and the density of the recognized epitope on the bacteria. Herein, we found substantial differences within the KD app/epitope density profiles in mice immunized with various species of heat-killed bacteria. The experiments further revealed a high cross-reactivity of the secreted IgG repertoires, binding to even unrelated bacteria with high affinity. This application confirmed the ability to quantify the anti-bacterial antibody repertoire and the utility of the developed bioassay to study the interplay between bacteria and the humoral response.
Collapse
Affiliation(s)
- Millie Heo
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Guilhem Chenon
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Carlos Castrillon
- Unit of Antibodies in Therapy and Pathology, Institute Pasteur, UMR1222 INSERM, F-75015, Paris, France
- 'Laboratoire de Biochimie' (LBC), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Jérôme Bibette
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Institute Pasteur, UMR1222 INSERM, F-75015, Paris, France
| | - Andrew D Griffiths
- 'Laboratoire de Biochimie' (LBC), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Jean Baudry
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France
| | - Klaus Eyer
- 'Laboratoire Colloïdes et Matériaux Divisés' (LCMD), ESPCI Paris, PSL Research University, CNRS UMR8231 Chimie Biologie Innovation, F-75005, Paris, France.
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, D-CHAB, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
29
|
Del Alcazar D, Wang Y, He C, Wendel BS, Del Río-Estrada PM, Lin J, Ablanedo-Terrazas Y, Malone MJ, Hernandez SM, Frank I, Naji A, Reyes-Terán G, Jiang N, Su LF. Mapping the Lineage Relationship between CXCR5 + and CXCR5 - CD4 + T Cells in HIV-Infected Human Lymph Nodes. Cell Rep 2020; 28:3047-3060.e7. [PMID: 31533030 PMCID: PMC6878759 DOI: 10.1016/j.celrep.2019.08.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/27/2019] [Accepted: 08/09/2019] [Indexed: 12/29/2022] Open
Abstract
CXCR5 is a key marker of follicular helper T (TFH) cells. Using primary lymph nodes (LNs) from HIV-infected patients, we identified a population of CXCR5− CD4+ T cells with TFH-cell-like features. This CXCR5− subset becomes expanded in severe HIV infection and is characterized by the upregulation of activation markers and high PD-1 and ICOS surface expression. Integrated analyses on the phenotypic heterogeneity, functional capacity, T cell receptor (TCR) repertoire, transcriptional profile, and epigenetic state of CXCR5−PD-1+ICOS+ T cells revealed a shared clonal relationship with TFH cells. CXCR5−PD-1+ICOS+ T cells retained a poised state for CXCR5 expression and exhibited a migratory transcriptional program. TCR sequence overlap revealed a contribution of LN-derived CXCR5−PD-1+ICOS+ T cells to circulating CXCR5− CD4+ T cells with B cell help function. These data link LN pathology to circulating T cells and expand the current understanding on the diversity of T cells that regulate B cell responses during chronic inflammation. Follicular helper T (TFH) cells are critical for antibody production. Del Alcazar et al. showed that TFH cells can lose their characteristic chemokine receptor, giving rise to migratory populations of CXCR5− T cells that retain B cell help function and are poised for CXCR5 expression.
Collapse
Affiliation(s)
- Daniel Del Alcazar
- Department of Medicine, Division of Rheumatology, Philadelphia VA Medical Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Yifeng Wang
- Department of Medicine, Division of Rheumatology, Philadelphia VA Medical Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chenfeng He
- Laboratory of Systems Immunology, Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Ben S Wendel
- Laboratory of Systems Immunology, Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA; McKetta Department of Chemical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Perla M Del Río-Estrada
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de México, México
| | - Jerome Lin
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuria Ablanedo-Terrazas
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de México, México
| | - Michael J Malone
- Laboratory of Systems Immunology, Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, College of Natural Sciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Stefany M Hernandez
- Laboratory of Systems Immunology, Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA; McKetta Department of Chemical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Ian Frank
- Department of Medicine, Division of Infectious Disease, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gustavo Reyes-Terán
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de México, México
| | - Ning Jiang
- Laboratory of Systems Immunology, Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, College of Natural Sciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura F Su
- Department of Medicine, Division of Rheumatology, Philadelphia VA Medical Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Eyer K, Castrillon C, Chenon G, Bibette J, Bruhns P, Griffiths AD, Baudry J. The Quantitative Assessment of the Secreted IgG Repertoire after Recall to Evaluate the Quality of Immunizations. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:1176-1184. [PMID: 32669311 PMCID: PMC7416324 DOI: 10.4049/jimmunol.2000112] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/15/2020] [Indexed: 01/03/2023]
Abstract
One of the major goals of vaccination is to prepare the body to rapidly secrete specific Abs during an infection. Assessment of the vaccine quality is often difficult to perform, as simple measurements like Ab titer only partly correlate with protection. Similarly, these simple measurements are not always sensitive to changes in the preceding immunization scheme. Therefore, we introduce in this paper a new, to our knowledge, method to assay the quality of immunization schemes for mice: shortly after a recall with pure Ag, we analyze the frequencies of IgG-secreting cells (IgG-SCs) in the spleen, as well as for each cells, the Ag affinity of the secreted Abs. We observed that after recall, appearance of the IgG-SCs within the spleen of immunized mice was fast (<24 h) and this early response was free of naive IgG-SCs. We further confirmed that our phenotypic analysis of IgG-SCs after recall strongly correlated with the different employed immunization schemes. Additionally, a phenotypic comparison of IgG-SCs presented in the spleen during immunization or after recall revealed similarities but also significant differences. The developed approach introduced a novel (to our knowledge), quantitative, and functional highly resolved alternative to study the quality of immunizations.
Collapse
Affiliation(s)
- Klaus Eyer
- Laboratoire Colloïdes et Matériaux Divisés, Institut Chimie, Biologie, Innovation, UMR8231, ESPCI Paris, CNRS, Université Paris Sciences et Lettres, 75005 Paris, France;
- Laboratory for Functional Immune Repertoire Analysis, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biology, ETH Zürich, 8093 Zurich, Switzerland
| | - Carlos Castrillon
- Unit of Antibodies in Therapy and Pathology, Pasteur Institute, UMR1222 INSERM, 75015 Paris, France
- Laboratoire de Biochimie, Institut Chimie, Biologie, Innovation, UMR8231, ESPCI Paris, CNRS, Université Paris Sciences et Lettres, 75005 Paris, France; and
- Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Guilhem Chenon
- Laboratoire Colloïdes et Matériaux Divisés, Institut Chimie, Biologie, Innovation, UMR8231, ESPCI Paris, CNRS, Université Paris Sciences et Lettres, 75005 Paris, France
| | - Jérôme Bibette
- Laboratoire Colloïdes et Matériaux Divisés, Institut Chimie, Biologie, Innovation, UMR8231, ESPCI Paris, CNRS, Université Paris Sciences et Lettres, 75005 Paris, France
| | - Pierre Bruhns
- Unit of Antibodies in Therapy and Pathology, Pasteur Institute, UMR1222 INSERM, 75015 Paris, France
| | - Andrew D Griffiths
- Laboratoire de Biochimie, Institut Chimie, Biologie, Innovation, UMR8231, ESPCI Paris, CNRS, Université Paris Sciences et Lettres, 75005 Paris, France; and
| | - Jean Baudry
- Laboratoire Colloïdes et Matériaux Divisés, Institut Chimie, Biologie, Innovation, UMR8231, ESPCI Paris, CNRS, Université Paris Sciences et Lettres, 75005 Paris, France
| |
Collapse
|
31
|
Detection of allergen-specific antibody-secreting cells in dogs by ELISPOT. Vet Immunol Immunopathol 2020; 228:110101. [PMID: 32861056 DOI: 10.1016/j.vetimm.2020.110101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 07/26/2020] [Accepted: 08/03/2020] [Indexed: 11/24/2022]
Abstract
Current laboratory tests are unable to distinguish healthy from allergic dogs. Unlike serum antibody responses, circulating antibody-secreting cells (ASC) are temporarily induced after each contact with the antigen. These ASC can be identified using ELISPOT and the observation of allergen-specific ASC might correlate with the causative allergens in dogs with an allergic dermatitis. In this study, blood was sampled from six privately-owned allergic dogs and six non-allergic laboratory beagles to determine the frequency of circulating allergen-specific ASC for common allergens. Blood IgE+, IgA + and IgG + cells were magnetically isolated to determine the number of allergen-specific ASC with ELISPOT for Dermatophagoides farinae, Dermatophagoides pteronyssinus, Alternaria alternata, birch, timothy grass, wheat, cow's milk, bovine, chicken and lamb meat. For IgA and IgG, allergen-specific spots were observed, however for IgE, no spots were detected for any of the allergens. ELISPOT could not differentiate allergic from non-allergic dogs. When the responses to the different allergens were compared, more IgA ASC for D. pteronyssinus were observed compared to some of the other allergens which was statistically significant for the non-allergic dogs and approached significance in the allergic dogs. These findings indicate that ELISPOT can be used to identify circulating allergen-specific IgA- and IgG-secreting cells. The technique did however not detect allergen-specific IgE ASC and was unable to distinguish allergic from non-allergic dogs. Only a small number of studies have studied allergen-specific IgA in dogs. The finding that dogs have higher numbers of D. pteronyssinus-specific IgA ASC points out that apart from IgE and IgG, it might be interesting to include IgA measurements for certain allergens to analyse the complete spectrum of both the protective and pro-allergic antibody responses.
Collapse
|
32
|
Pedreño-Lopez N, Ricciardi MJ, Rosen BC, Song G, Andrabi R, Burton DR, Rakasz EG, Watkins DI. An Automated Fluorescence-Based Method to Isolate Bone Marrow-Derived Plasma Cells from Rhesus Macaques Using SIVmac239 SOSIP.664. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:781-790. [PMID: 32953929 PMCID: PMC7476808 DOI: 10.1016/j.omtm.2020.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/31/2020] [Indexed: 11/20/2022]
Abstract
Simian immunodeficiency virus (SIV) infection of Indian rhesus macaques (RMs) is one of the best-characterized animal models for human immunodeficiency virus (HIV) infection. Monoclonal antibodies (mAbs) have shown promise for prevention and treatment of HIV infection. However, it has been difficult to isolate mAbs that potently neutralize the highly pathogenic SIVmac239 strain. This has been largely due to the low frequency of circulating B cells encoding neutralizing Abs. Here we describe a novel technique to isolate mAbs directly from bone marrow-derived, Ab-secreting plasma cells. We employed an automated micromanipulator to isolate single SIVmac239 SOSIP.664-specific plasma cells from the bone marrow of a SIVmac239-infected RM with serum neutralization titers against SIVmac239. After picking plasma cells, we obtained 44 paired Ab sequences. Ten of these mAbs were SIV specific. Although none of these mAbs neutralized SIVmac239, three mAbs completely neutralized the related SIVmac316 strain. The majority of these mAbs bound to primary rhesus CD4+ T cells infected with SIVmac239 and induced Ab-dependent cellular cytotoxicity. This method is a first step in successful isolation of antigen-specific bone marrow-derived plasma cells from RMs.
Collapse
Affiliation(s)
- Nuria Pedreño-Lopez
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
- Corresponding author: Nuria Pedreño-Lopez, Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA.
| | - Michael J. Ricciardi
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Brandon C. Rosen
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
- Medical Scientist Training Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - David I. Watkins
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
33
|
Meyer Sauteur PM, Seiler M, Trück J, Unger WWJ, Paioni P, Relly C, Staubli G, Haas T, Gysin C, M Bachmann L, van Rossum AMC, Berger C. Diagnosis of Mycoplasma pneumoniae Pneumonia with Measurement of Specific Antibody-Secreting Cells. Am J Respir Crit Care Med 2020; 200:1066-1069. [PMID: 31251669 PMCID: PMC6794114 DOI: 10.1164/rccm.201904-0860le] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | | | - Johannes Trück
- University Children's Hospital ZurichZurich, Switzerland
| | - Wendy W J Unger
- Erasmus MC University Medical Center-Sophia Children's HospitalRotterdam, the Netherlandsand
| | - Paolo Paioni
- University Children's Hospital ZurichZurich, Switzerland
| | - Christa Relly
- University Children's Hospital ZurichZurich, Switzerland
| | - Georg Staubli
- University Children's Hospital ZurichZurich, Switzerland
| | - Thorsten Haas
- University Children's Hospital ZurichZurich, Switzerland
| | - Claudine Gysin
- University Children's Hospital ZurichZurich, Switzerland
| | | | | | | |
Collapse
|
34
|
Pedreño-Lopez N, Dang CM, Rosen BC, Ricciardi MJ, Bailey VK, Gutman MJ, Gonzalez-Nieto L, Pauthner MG, Le K, Song G, Andrabi R, Weisgrau KL, Pomplun N, Martinez-Navio JM, Fuchs SP, Wrammert J, Rakasz EG, Lifson JD, Martins MA, Burton DR, Watkins DI, Magnani DM. Induction of Transient Virus Replication Facilitates Antigen-Independent Isolation of SIV-Specific Monoclonal Antibodies. Mol Ther Methods Clin Dev 2020; 16:225-237. [PMID: 32083148 PMCID: PMC7021589 DOI: 10.1016/j.omtm.2020.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/26/2020] [Indexed: 02/04/2023]
Abstract
Structural characterization of the HIV-1 Envelope (Env) glycoprotein has facilitated the development of Env probes to isolate HIV-specific monoclonal antibodies (mAbs). However, preclinical studies have largely evaluated these virus-specific mAbs against chimeric viruses, which do not naturally infect non-human primates, in contrast to the unconstrained simian immunodeficiency virus (SIV)mac239 clone. Given the paucity of native-like reagents for the isolation of SIV-specific B cells, we examined a method to isolate SIVmac239-specific mAbs without using Env probes. We first activated virus-specific B cells by inducing viral replication after the infusion of a CD8β-depleting mAb or withdrawal of antiretroviral therapy in SIVmac239-infected rhesus macaques. Following the rise in viremia, we observed 2- to 4-fold increases in the number of SIVmac239 Env-reactive plasmablasts in circulation. We then sorted these activated B cells and obtained 206 paired Ab sequences. After expressing 122 mAbs, we identified 14 Env-specific mAbs. While these Env-specific mAbs bound to both the SIVmac239 SOSIP.664 trimer and to infected primary rhesus CD4+ T cells, five also neutralized SIVmac316. Unfortunately, none of these mAbs neutralized SIVmac239. Our data show that this method can be used to isolate virus-specific mAbs without antigenic probes by inducing bursts of contemporary replicating viruses in vivo.
Collapse
Affiliation(s)
- Nuria Pedreño-Lopez
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Christine M. Dang
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Brandon C. Rosen
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
- Medical Scientist Training Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Michael J. Ricciardi
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Varian K. Bailey
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Martin J. Gutman
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Lucas Gonzalez-Nieto
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Matthias G. Pauthner
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (Scripps CHAVI-ID), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Khoa Le
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (Scripps CHAVI-ID), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kim L. Weisgrau
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Nicholas Pomplun
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - José M. Martinez-Navio
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Sebastian P. Fuchs
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Jens Wrammert
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30317, USA
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Mauricio A. Martins
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (Scripps CHAVI-ID), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - David I. Watkins
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Diogo M. Magnani
- Department of Pathology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
35
|
Ascough S, Vlachantoni I, Kalyan M, Haijema BJ, Wallin-Weber S, Dijkstra-Tiekstra M, Ahmed MS, van Roosmalen M, Grimaldi R, Zhang Q, Leenhouts K, Openshaw PJ, Chiu C. Local and Systemic Immunity against Respiratory Syncytial Virus Induced by a Novel Intranasal Vaccine. A Randomized, Double-Blind, Placebo-controlled Clinical Trial. Am J Respir Crit Care Med 2020; 200:481-492. [PMID: 30753101 DOI: 10.1164/rccm.201810-1921oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Rationale: Needle-free intranasal vaccines offer major potential advantages, especially against pathogens entering via mucosal surfaces. As yet, there is no effective vaccine against respiratory syncytial virus (RSV), a ubiquitous pathogen of global importance that preferentially infects respiratory epithelial cells; new strategies are urgently required.Objectives: Here, we report the safety and immunogenicity of a novel mucosal RSV F protein vaccine linked to an immunostimulatory bacterium-like particle (BLP).Methods: In this phase I, randomized, double-blind, placebo-controlled trial, 48 healthy volunteers, aged 18-49 years, were randomly assigned to receive placebo or SynGEM (low or high dose) intranasally by prime-boost administration. The primary outcome was safety and tolerability, with secondary objectives assessing virus-specific immunogenicity.Measurements and Main Results: There were no significant differences in adverse events between placebo and vaccinated groups. SynGEM induced systemic plasmablast responses and significant, durable increases in RSV-specific serum antibody in healthy, seropositive adults. Volunteers given low-dose SynGEM (140 μg F, 2 mg BLP) required a boost at Day 28 to achieve plateau responses with a maximum fold change of 2.4, whereas high-dose recipients (350 μg F, 5 mg BLP) achieved plateau responses with a fold change of 1.5 after first vaccination that remained elevated up to 180 days after vaccination, irrespective of further boosting. Palivizumab-like antibodies were consistently induced, but F protein site ∅-specific antibodies were not detected, and virus-specific nasal IgA responses were heterogeneous, with the strongest responses in individuals with lower pre-existing antibody levels.Conclusions: SynGEM is thus the first nonreplicating intranasal RSV subunit vaccine to induce persistent antibody responses in human volunteers.Clinical trials registered with www.clinicaltrials.gov (NCT02958540).
Collapse
Affiliation(s)
- Stephanie Ascough
- 1Section of Infectious Diseases and Immunity, Department of Medicine, and.,2Section of Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Iris Vlachantoni
- 2Section of Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Mohini Kalyan
- 1Section of Infectious Diseases and Immunity, Department of Medicine, and.,2Section of Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Bert-Jan Haijema
- 3Mucosis B.V., represented by trustee Mr. Holtz, LLM, Bout Advocaten, Groningen, and Virtuvax B.V., Groningen, the Netherlands; and
| | - Sanna Wallin-Weber
- 3Mucosis B.V., represented by trustee Mr. Holtz, LLM, Bout Advocaten, Groningen, and Virtuvax B.V., Groningen, the Netherlands; and
| | - Margriet Dijkstra-Tiekstra
- 3Mucosis B.V., represented by trustee Mr. Holtz, LLM, Bout Advocaten, Groningen, and Virtuvax B.V., Groningen, the Netherlands; and
| | - Muhammad S Ahmed
- 4Department of Clinical Infection, Microbiology, and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Maarten van Roosmalen
- 3Mucosis B.V., represented by trustee Mr. Holtz, LLM, Bout Advocaten, Groningen, and Virtuvax B.V., Groningen, the Netherlands; and
| | - Roberto Grimaldi
- 3Mucosis B.V., represented by trustee Mr. Holtz, LLM, Bout Advocaten, Groningen, and Virtuvax B.V., Groningen, the Netherlands; and
| | - Qibo Zhang
- 4Department of Clinical Infection, Microbiology, and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Kees Leenhouts
- 3Mucosis B.V., represented by trustee Mr. Holtz, LLM, Bout Advocaten, Groningen, and Virtuvax B.V., Groningen, the Netherlands; and
| | - Peter J Openshaw
- 2Section of Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Christopher Chiu
- 1Section of Infectious Diseases and Immunity, Department of Medicine, and
| |
Collapse
|
36
|
Booster vaccination with a fractional dose of an oral cholera vaccine induces comparable vaccine-specific antibody avidity as a full dose: A randomised clinical trial. Vaccine 2020; 38:655-662. [DOI: 10.1016/j.vaccine.2019.10.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 01/06/2023]
|
37
|
Gaultier GN, McCready W, Ulanova M. The effect of pneumococcal immunization on total and antigen-specific B cells in patients with severe chronic kidney disease. BMC Immunol 2019; 20:41. [PMID: 31718534 PMCID: PMC6849264 DOI: 10.1186/s12865-019-0325-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022] Open
Abstract
Background While the 23-valent pneumococcal polysaccharide vaccine (PPV23) is routinely used in Canada and some other countries to prevent pneumococcal infection in adults with chronic kidney disease (CKD), patients develop a suboptimal antibody response to PPV23 due to their immune dysfunction. The 13-valent pneumococcal conjugate vaccine (PCV13) has superior immunogenicity in some categories of immunocompromised adults; however, its effect on the immune response in CKD patients has only been addressed by two recent studies with conflicting results. The effect of PPV23 or PCV13 on B cells in these patients has not been previously studied. We studied the absolute numbers and proportions of B cells and subpopulations in two groups of adult patients with severe CKD pre- and 7 days post-immunization with PCV13: pneumococcal vaccine naïve and previously immunized with PPV23 (over one year ago). Results PPV23 immunized patients had significantly lower proportions and absolute numbers of class switched memory (CD19 + CD27 + IgM-), as well as lower absolute numbers of IgM memory (CD19 + CD27 + IgM+) and class switched B cells (CD19 + CD27-IgM-) compared to PPV23 naïve patients. Following PCV13 immunization, the differences in absolute numbers of B-cell subpopulations between groups remained significant. The PPV23 immunized group had higher proportions of CD5- B cells along with lower proportions and absolute numbers of CD5+ B cells compared to PPV23 naïve patients both pre- and post-immunization with PCV13. However, previous PPV23 immunization did not have a noticeable effect on the numbers of total IgG or serotype 6B and 14 specific antibody-secreting cells detected 7 days post-immunization with PCV13. Nevertheless, fold increase in anti-serotype 14 IgG concentrations 28 days post-PCV13 was greater in PPV23 naïve than in previously immunized patients. Conclusions The results suggest that immunization with PPV23 may result in long-term changes in B-cell subpopulations such as increased prevalence of CD5- B cells and decreased prevalence of class switched memory B cells in the peripheral blood. Because previous immunization with PPV23 in patients with CKD is associated with a significant decrease in the total class switched memory B cells in response to subsequent immunization with PCV13, this may reduce PCV13 immunogenicity in the setting of PPV23 followed by PCV13. Trial registration Registered February 24, 2015 at ClinicalTrials.gov (NCT 02370069).
Collapse
Affiliation(s)
| | - William McCready
- Division of Medical Sciences, Northern Ontario School of Medicine, Lakehead University, 955 Oliver Road, Thunder Bay, ON, P7B 5E1, Canada
| | - Marina Ulanova
- Department of Biology, Lakehead University, Thunder Bay, Canada. .,Division of Medical Sciences, Northern Ontario School of Medicine, Lakehead University, 955 Oliver Road, Thunder Bay, ON, P7B 5E1, Canada.
| |
Collapse
|
38
|
Shi B, Wu Y, Wang C, Li X, Yu F, Wang B, Yang Z, Li J, Liang M, Wen Y, Ying T, Yuan Z. Evaluation of antiviral - passive - active immunization ("sandwich") therapeutic strategy for functional cure of chronic hepatitis B in mice. EBioMedicine 2019; 49:247-257. [PMID: 31680000 PMCID: PMC6945269 DOI: 10.1016/j.ebiom.2019.10.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/14/2019] [Accepted: 10/23/2019] [Indexed: 01/14/2023] Open
Abstract
Background Chronic Hepatitis B (CHB) remains a major problem for global public health. Viral persistence and immune defects are the two major reasons for CHB, and it was hypothesized that based on a transient clearance of serum viral DNA and HBsAg “window stage”, active immunization against hepatitis B virus (HBV) might initiate effective host immune responses versus HBV to achieve functional cure of CHB. Methods Two experimental mouse models that mice hydrodynamic injected HBV DNA or infected with recombinant AAV/HBV were used. The “sandwich” therapeutic effect by using a potent human anti-HBsAg neutralizing monoclonal antibody (G12) in combination with antiviral drug tenofovir disoproxil fumarate (TDF), followed by active immunization with HBsAg-HBsAb (mYIC) was evaluated. Findings A single G12 injection rapidly cleared serum HBsAg in HDI-HBV carrier mice, with a synergistic effect in decreasing viral DNA load when TDF was given orally. When both serum viral DNA and HBsAg load became low or undetectable, mYIC was administered. A more effective clearance of viral DNA and HBsAg was observed and serum HBsAb was developed only in these “sandwich”-treated mice. Efficient intrahepatic anti-HBV immune responses were also observed in these mice, including the formation of aggregates of myeloid cells with CD8+T cells and increased TNF-α, granzyme B production. Interpretation The “sandwich” combination therapy not only efficiently decreased HBsAg and HBV DNA levels but also induced effective cellular and humoral immunity, which may result in functional cure of CHB.
Collapse
Affiliation(s)
- Bisheng Shi
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China; Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Rd, Jinshan District, Shanghai, PR China
| | - Yanling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China
| | - Chunyu Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China
| | - Xiaofang Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China
| | - Fan Yu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China
| | - Bin Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China
| | - Zhenlin Yang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China; Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Xuhui District, Shanghai, PR China
| | - Jianhua Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China
| | - Mifang Liang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, PR China
| | - Yumei Wen
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China.
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China.
| | - Zhenghong Yuan
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, 138 Dong'an Rd, Xuhui District, Shanghai, PR China.
| |
Collapse
|
39
|
Gonzalez-Nieto L, Castro IM, Bischof GF, Shin YC, Ricciardi MJ, Bailey VK, Dang CM, Pedreño-Lopez N, Magnani DM, Ejima K, Allison DB, Gil HM, Evans DT, Rakasz EG, Lifson JD, Desrosiers RC, Martins MA. Vaccine protection against rectal acquisition of SIVmac239 in rhesus macaques. PLoS Pathog 2019; 15:e1008015. [PMID: 31568531 PMCID: PMC6791558 DOI: 10.1371/journal.ppat.1008015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/14/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023] Open
Abstract
A prophylactic vaccine against human immunodeficiency virus (HIV) remains a top priority in biomedical research. Given the failure of conventional immunization protocols to confer robust protection against HIV, new and unconventional approaches may be needed to generate protective anti-HIV immunity. Here we vaccinated rhesus macaques (RMs) with a recombinant (r)DNA prime (without any exogenous adjuvant), followed by a booster with rhesus monkey rhadinovirus (RRV)-a herpesvirus that establishes persistent infection in RMs (Group 1). Both the rDNA and rRRV vectors encoded a near-full-length simian immunodeficiency virus (SIVnfl) genome that assembles noninfectious SIV particles and expresses all nine SIV gene products. This rDNA/rRRV-SIVnfl vaccine regimen induced persistent anti-Env antibodies and CD8+ T-cell responses against the entire SIV proteome. Vaccine efficacy was assessed by repeated, marginal-dose, intrarectal challenges with SIVmac239. Encouragingly, vaccinees in Group 1 acquired SIVmac239 infection at a significantly delayed rate compared to unvaccinated controls (Group 3). In an attempt to improve upon this outcome, a separate group of rDNA/rRRV-SIVnfl-vaccinated RMs (Group 2) was treated with a cytotoxic T-lymphocyte antigen-4 (CTLA-4)-blocking monoclonal antibody during the vaccine phase and then challenged in parallel with Groups 1 and 3. Surprisingly, Group 2 was not significantly protected against SIVmac239 infection. In sum, SIVnfl vaccination can protect RMs against rigorous mucosal challenges with SIVmac239, a feat that until now had only been accomplished by live-attenuated strains of SIV. Further work is needed to identify the minimal requirements for this protection and whether SIVnfl vaccine efficacy can be improved by means other than anti-CTLA-4 adjuvant therapy.
Collapse
Affiliation(s)
- Lucas Gonzalez-Nieto
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Isabelle M. Castro
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Georg F. Bischof
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Young C. Shin
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Michael J. Ricciardi
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Varian K. Bailey
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Christine M. Dang
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Nuria Pedreño-Lopez
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Diogo M. Magnani
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Keisuke Ejima
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, United States of America
| | - David B. Allison
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, United States of America
| | - Hwi Min Gil
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - David T. Evans
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Ronald C. Desrosiers
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| | - Mauricio A. Martins
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, United States of America
| |
Collapse
|
40
|
Boonyaratanakornkit J, Taylor JJ. Techniques to Study Antigen-Specific B Cell Responses. Front Immunol 2019; 10:1694. [PMID: 31396218 PMCID: PMC6667631 DOI: 10.3389/fimmu.2019.01694] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
Antibodies against foreign antigens are a critical component of the overall immune response and can facilitate pathogen clearance during a primary infection and also protect against subsequent infections. Dysregulation of the antibody response can lead to an autoimmune disease, malignancy, or enhanced infection. Since the experimental delineation of a distinct B cell lineage in 1965, various methods have been developed to understand antigen-specific B cell responses in the context of autoimmune diseases, primary immunodeficiencies, infection, and vaccination. In this review, we summarize the established techniques and discuss new and emerging technologies for probing the B cell response in vitro and in vivo by taking advantage of the specificity of B cell receptor (BCR)-associated and secreted antibodies. These include ELISPOT, flow cytometry, mass cytometry, and fluorescence microscopy to identify and/or isolate primary antigen-specific B cells. We also present our approach to identify rare antigen-specific B cells using magnetic enrichment followed by flow cytometry. Once these cells are isolated, in vitro proliferation assays and adoptive transfer experiments in mice can be used to further characterize antigen-specific B cell activation, function, and fate. Transgenic mouse models of B cells targeting model antigens and of B cell signaling have also significantly advanced our understanding of antigen-specific B cell responses in vivo.
Collapse
Affiliation(s)
- Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | | |
Collapse
|
41
|
Park KS. Nucleic acid aptamer-based methods for diagnosis of infections. Biosens Bioelectron 2018; 102:179-188. [PMID: 29136589 PMCID: PMC7125563 DOI: 10.1016/j.bios.2017.11.028] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/20/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
Infectious diseases are a serious global problem, which not only take an enormous human toll but also incur tremendous economic losses. In combating infectious diseases, rapid and accurate diagnostic tests are required for pathogen identification at the point of care (POC). In this review, investigations of diagnostic strategies for infectious diseases that are based on aptamers, especially nucleic acid aptamers, oligonucleotides that have high affinities and specificities toward their targets, are described. Owing to their unique features including low cost of production, easy chemical modification, high chemical stability, reproducibility, and low levels of immunogenicity and toxicity, aptamers have been widely utilized as bio-recognition elements (bio-receptors) for the development of infection diagnostic systems. We discuss nucleic acid aptamer-based methods that have been developed for diagnosis of infections using a format that organizes discussion according to the target pathogenic analytes including toxins or proteins, whole cells and nucleic acids. Also included is, a summary of recent advances made in the sensitive detection of pathogenic bacteria utilizing the isothermal nucleic acid amplification method. Lastly, a nucleic acid aptamer-based POC system is described and future directions of studies in this area are discussed.
Collapse
Affiliation(s)
- Ki Soo Park
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
42
|
Cárdeno A, Magnusson MK, Quiding-Järbrink M, Lundgren A. Activated T follicular helper-like cells are released into blood after oral vaccination and correlate with vaccine specific mucosal B-cell memory. Sci Rep 2018; 8:2729. [PMID: 29426881 PMCID: PMC5807513 DOI: 10.1038/s41598-018-20740-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
T follicular helper (Tfh)-like cells with potent B-cell helping ability are mobilized into human circulation after parenteral vaccination and are generally held to reflect ongoing germinal center reactions. However, whether mucosal vaccination induces systemic Tfh responses and how such responses may relate to IgA production are unknown. We investigated the frequencies, phenotype and function of circulating Tfh-like CD4+CXCR5+ T cells (cTfh) in adults receiving an oral inactivated enterotoxigenic Escherichia coli vaccine. Subjects were classified as vaccine responders or weak/non-responders based on their intestine-derived antibody-secreting cell (ASC) IgA responses to major vaccine antigens. Oral immunization induced significantly increased proportions of cTfh cells expressing the cTfh activation marker inducible costimulator (ICOS) in ASC responders, but not in weak/non-responders. Vaccination also enhanced the expression of IL-21, Th17 markers and integrin β7 by activated cTfh cells, supporting functionality and gut homing potential. cTfh cells promoted total and vaccine specific IgA production from cocultured B cells. Magnitudes of cTfh responses assessed within a week after primary vaccinations correlated with memory intestine-derived vaccine specific IgA responses 1-2 years later. We conclude that activated ICOS+ Tfh-like cells are mobilized into blood after oral vaccination and may be used as biomarkers of vaccine specific mucosal memory in humans.
Collapse
Affiliation(s)
- Ana Cárdeno
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Maria K Magnusson
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | | | - Anna Lundgren
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
43
|
Sinha A, Kanungo S, Kim DR, Manna B, Song M, Park JY, Haldar B, Sharma P, Mallick AH, Kim SA, Babji S, Sur D, Kang G, Ali M, Petri WA, Wierzba TF, Czerkinsky C, Nandy RK, Dey A. Antibody secreting B cells and plasma antibody response to rotavirus vaccination in infants from Kolkata India. Heliyon 2018; 4:e00519. [PMID: 29560435 PMCID: PMC5857522 DOI: 10.1016/j.heliyon.2018.e00519] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 01/28/2023] Open
Abstract
Background Assessing immune response after rotavirus vaccination consists in measuring serum or plasma IgA and IgG antibodies, but these assays provide very little information about the mucosal immune response. Thus the development of assays for detection of mucosal immune response following rotavirus vaccination is essential. We evaluate to assess circulating antibody-secreting cells (ASCs) as a potential means to evaluate mucosal immune responses to rotavirus vaccine. Methods 372 subjects, aged 6 weeks, were enrolled in the study. All the subjects were assigned to receive two doses of Rotarix® vaccine. Using a micro-modified whole blood-based ELISPOT assay, circulating rotavirus type-specific IgA- and IgG-ASCs, including gut homing β7+ ASCs, were enumerated on week 6 before the first dose of Rotarix vaccination at 7 weeks of age and week 18 after the second vaccination at 17 weeks of age. Plasma samples collected before vaccination, and after two doses of Rotarix® vaccination were tested for plasma rotavirus IgA titers. Results Two doses of Rotarix® provided to induce sero-protective titer of ≥ 20 Units in 35% of subjects. Total blood IgA- ASC responses were detected in 26.4% of subjects who were non-responder before vaccination. Among responders, 47% of the subjects also have sero-protective plasma IgA titers. Discussion Our results suggest that virus-specific blood gut homing ASCs were detected and provide insight into mucosal immune response after rotavirus vaccination. Further studies are needed to evaluate the duration of such immune responses and to assess the programmatic utility of this whole blood-based mucosal ASC testing for the rotavirus immunization program.
Collapse
Affiliation(s)
- Anuradha Sinha
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Suman Kanungo
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | | | - Byomkesh Manna
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Manki Song
- International Vaccine Institute, Seoul, South Korea
| | - Ju Yeon Park
- International Vaccine Institute, Seoul, South Korea
| | - Bisakha Haldar
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Prashant Sharma
- Department of Microbiology and Immunology, Seoul National University
| | | | - Soon Ae Kim
- International Vaccine Institute, Seoul, South Korea
| | - Sudhir Babji
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Dipika Sur
- National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Gagandeep Kang
- Division of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Mohammad Ali
- Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | | | | | - Cecil Czerkinsky
- Institut de Pharmacologie Moleculaire & Cellulaire, CNRS-INSERM-University of Nice-Sophia Antipolis, Valbonne, France
| | | | - Ayan Dey
- International Vaccine Institute, Seoul, South Korea
| |
Collapse
|
44
|
Hwai H, Chen YY, Tzeng SJ. B-Cell ELISpot Assay to Quantify Antigen-Specific Antibody-Secreting Cells in Human Peripheral Blood Mononuclear Cells. Methods Mol Biol 2018; 1808:133-141. [PMID: 29956179 DOI: 10.1007/978-1-4939-8567-8_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Peripheral blood is commonly used to assess the cellular and humoral immune responses in clinical studies. It is a convenient sample to collect for immunological research as compared to the surgically excised and biopsied lymphoid specimens. To determine the functional status of immune system from peripheral blood, the enzyme-linked immunospot (ELISpot) assay is a popular method of choice owing to its high sensitivity, great accuracy, and easy performance. The ELISpot allows detection and quantification of cellular functionality at the single-cell level. Therefore, ELISpot assay is commonly applied to detect cytokines and cytotoxic granules released from T cells as well as to measure antibodies secreted from B cells. Because the ELISpot assay has been increasingly used for evaluation of the vaccine efficacy in clinical trials, standardization and reproducibility are crucial to minimize assay variability amongst samples from different sources. Here we introduce methods to isolate human peripheral blood mononuclear cells (PBMCs) for quantification of the antigen-specific antibody-secreting cells using the ELISpot assay.
Collapse
Affiliation(s)
- Haw Hwai
- Department of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Ying Chen
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shiang-Jong Tzeng
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
45
|
Agrawal L, Engel KB, Greytak SR, Moore HM. Understanding preanalytical variables and their effects on clinical biomarkers of oncology and immunotherapy. Semin Cancer Biol 2017; 52:26-38. [PMID: 29258857 DOI: 10.1016/j.semcancer.2017.12.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 12/20/2022]
Abstract
Identifying a suitable course of immunotherapy treatment for a given patient as well as monitoring treatment response is heavily reliant on biomarkers detected and quantified in blood and tissue biospecimens. Suboptimal or variable biospecimen collection, processing, and storage practices have the potential to alter clinically relevant biomarkers, including those used in cancer immunotherapy. In the present review, we summarize effects reported for immunologically relevant biomarkers and highlight preanalytical factors associated with specific analytical platforms and assays used to predict and gauge immunotherapy response. Given that many of the effects introduced by preanalytical variability are gene-, transcript-, and protein-specific, biospecimen practices should be standardized and validated for each biomarker and assay to ensure accurate results and facilitate clinical implementation of newly identified immunotherapy approaches.
Collapse
Affiliation(s)
- Lokesh Agrawal
- Biorepositories and Biospecimen Research Branch (BBRB), Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Drive, Bethesda, Maryland, USA
| | | | | | - Helen M Moore
- Biorepositories and Biospecimen Research Branch (BBRB), Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, 9609 Medical Center Drive, Bethesda, Maryland, USA.
| |
Collapse
|
46
|
Park KS, Kim H, Kim S, Lee K, Park S, Song J, Min C, Khanam F, Rashu R, Bhuiyan TR, Ryan ET, Qadri F, Weissleder R, Cheon J, Charles RC, Lee H. Nanomagnetic System for Rapid Diagnosis of Acute Infection. ACS NANO 2017; 11:11425-11432. [PMID: 29121461 PMCID: PMC6296367 DOI: 10.1021/acsnano.7b06074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Pathogen-activated antibody-secreting cells (ASCs) produce and secrete antigen-specific antibodies. ASCs are detectable in the peripheral blood as early as 3 days after antigen exposure, which makes ASCs a potential biomarker for early disease detection. Here, we present a magnetic capture and detection (MCD) assay for sensitive, on-site detection of ASCs. In this approach, ASCs are enriched through magnetic capture, and secreted antibodies are magnetically detected by a miniaturized nuclear magnetic resonance (μNMR) system. This approach is based entirely on magnetics, which supports high contrast against biological background and simplifies assay procedures. We advanced the MCD system by (i) synthesizing magnetic nanoparticles with high magnetic moments for both cell capture and antibody detection, (ii) developing a miniaturized magnetic device for high-yield cell capture, and (iii) optimizing the μNMR assay for antibody detection. Antibody responses targeting hemolysin E (HlyE) can accurately identify individuals with acute enteric fever. As a proof-of-concept, we applied MCD to detect antibodies produced by HlyE-specific hybridoma cells. The MCD achieved high sensitivity in detecting antibodies secreted from as few as 5 hybridoma cells (50 cells/mL). Importantly, the assay could be performed with whole blood with minimal sample processing.
Collapse
Affiliation(s)
- Ki Soo Park
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Center for NanoMedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Hoyoung Kim
- Center for NanoMedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Soojin Kim
- Center for NanoMedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyungheon Lee
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Sohyeon Park
- Center for NanoMedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jun Song
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Changwook Min
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Farhana Khanam
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Rasheduzzaman Rashu
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Edward T. Ryan
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jinwoo Cheon
- Center for NanoMedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Richelle C. Charles
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Center for NanoMedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
47
|
Assessing antigen specific HLA-DR+ antibody secreting cell (DR+ASC) responses in whole blood in enteric infections using an ELISPOT technique. Microbes Infect 2017; 20:122-129. [PMID: 29104143 DOI: 10.1016/j.micinf.2017.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 11/23/2022]
Abstract
Antibody secreting cells (ASCs) generate antibodies in an antigen-specific manner as part of the adaptive immune response to infections, and these cells increase their surface expression of HLA-DR. We have studied this parameter (HLA-DR+ ASC) in patients with recent diarrheal infection using immuno-magnetic cell sorting and an enzyme linked immunospot (ELISPOT) technique that requires only one milliliter of blood. We validated this approach in adult patients with cholera (n = 15) or ETEC diarrhea (n = 30) on days 2, 7 and 30 after showing clinical symptom at the International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) hospital in Dhaka, and we compared responses to age-matched healthy controls (n = 7). We found that HLA-DR+ ASC (DR+ASC) responses specific both for T cell-dependent (cholera toxin B subunit), and T cell-independent (lipopolysaccharide) antigens were elevated at day 7 after showing clinical cholera symptom. Similarly, DR+ASCs were elevated against both heat-labile toxin and colonization factors following ETEC infection. We observed significant correlations between antigen-specific DR+ASC responses and antigen-specific, gut homing ASC and plasma antibody responses. This study demonstrates that a simple ELISPOT procedure allows determination of antigen-specific ASC responses using a small volume of whole blood following diarrhea. This technique may be particularly useful in studying DR+ASC responses in young children and infants, either following infection or vaccination.
Collapse
|
48
|
Charles RC, Nakajima R, Liang L, Jasinskas A, Berger A, Leung DT, Kelly M, Xu P, Kovác P, Giffen SR, Harbison JD, Chowdhury F, Khan AI, Calderwood SB, Bhuiyan TR, Harris JB, Felgner PL, Qadri F, Ryan ET. Plasma and Mucosal Immunoglobulin M, Immunoglobulin A, and Immunoglobulin G Responses to the Vibrio cholerae O1 Protein Immunome in Adults With Cholera in Bangladesh. J Infect Dis 2017; 216:125-134. [PMID: 28535267 PMCID: PMC5853614 DOI: 10.1093/infdis/jix253] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/19/2017] [Indexed: 11/16/2022] Open
Abstract
Background. Cholera is a severe dehydrating illness of humans caused by toxigenic strains of Vibrio cholerae O1 or O139. Identification of immunogenic V. cholerae antigens could lead to a better understanding of protective immunity in human cholera. Methods. We probed microarrays containing 3652 V. cholerae antigens with plasma and antibody-in-lymphocyte supernatant (ALS, a surrogate marker of mucosal immune responses) from patients with severe cholera caused by V. cholerae O1 in Bangladesh and age-, sex-, and ABO-matched Bangladeshi controls. We validated a subset of identified antigens using enzyme-linked immunosorbent assay. Results. Overall, we identified 608 immunoreactive V. cholerae antigens in our screening, 59 of which had higher immunoreactivity in convalescent compared with acute-stage or healthy control samples (34 in plasma, 39 in mucosal ALS; 13 in both sample sets). Identified antigens included cholera toxin B and A subunits, V. cholerae O–specific polysaccharide and lipopolysaccharide, toxin coregulated pilus A, sialidase, hemolysin A, flagellins (FlaB, FlaC, and FlaD), phosphoenolpyruvate-protein phosphotransferase, and diaminobutyrate–2-oxoglutarate aminotransferase. Conclusions. This study is the first antibody profiling of the mucosal and systemic antibody responses to the nearly complete V. cholerae O1 protein immunome; it has identified antigens that may aid in the development of an improved cholera vaccine.
Collapse
Affiliation(s)
- Richelle C Charles
- Division of Infectious Diseases, Massachusetts General Hospital.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Rie Nakajima
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine
| | - Li Liang
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine
| | - Al Jasinskas
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine
| | - Amanda Berger
- Division of Infectious Diseases, Massachusetts General Hospital
| | - Daniel T Leung
- Division of Infectious Diseases, Department of Medicine, University of Utah School of Medicine, Salt Lake City
| | - Meagan Kelly
- Division of Infectious Diseases, Massachusetts General Hospital
| | - Peng Xu
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Pavol Kovác
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Samantha R Giffen
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Fahima Chowdhury
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka
| | - Ashraful I Khan
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka
| | - Stephen B Calderwood
- Division of Infectious Diseases, Massachusetts General Hospital.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | | | - Jason B Harris
- Division of Infectious Diseases, Massachusetts General Hospital.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Philip L Felgner
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka
| | - Edward T Ryan
- Division of Infectious Diseases, Massachusetts General Hospital.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
49
|
Single-cell deep phenotyping of IgG-secreting cells for high-resolution immune monitoring. Nat Biotechnol 2017; 35:977-982. [DOI: 10.1038/nbt.3964] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 08/17/2017] [Indexed: 01/04/2023]
|
50
|
A recombinant protein of Salmonella Typhi induces humoral and cell-mediated immune responses including memory responses. Vaccine 2017; 35:4523-4531. [PMID: 28739115 DOI: 10.1016/j.vaccine.2017.07.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/27/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022]
Abstract
Gram negative enteric bacteria, Salmonella enterica serovar Typhi (S. Typhi), the etiological agent of typhoid fever is a major public health problem in developing countries. While a permanent solution to the problem would require improved sanitation, food and water hygiene, controlling the infection by vaccination is urgently required due to the emergence of multidrug resistant strains in multiple countries. The currently licensed vaccines are moderately efficacious with limited applicability, and no recommended vaccines exist for younger children. We had previously reported that a candidate vaccine based on recombinant outer membrane protein (rT2544) of S. Typhi is highly immunogenic and protective in mice. Here we show that rT2544-specific antiserum is capable of mediating bacterial lysis by the splenocytes through Antibody-Dependent Cellular Cytotoxicity (ADCC). Increased populations of rT2544-specific IgA and IgG secreting plasma cells are found in the spleen, mesenteric lymph nodes and peyer's patches. Cell-Mediated Immune Responses (CMIR) induced by rT2544 consist of Th1 cell differentiation and generation of cytotoxic T lymphocytes (CTL), which produce IFN-γ and are capable of destroying cells displaying T2544-derived antigens. rT2544 elicits pro-inflammatory cytokines (TNF-α, IL-6) from Bone Marrow-Derived Dendritic cells (BMDCs), while in vitro re-stimulation of rT2544-primed CD4+ T cells induces cell proliferation and generates higher amounts of Th1 cytokines, such as IFN-gamma, TNF-α and IL-2. Finally, the candidate vaccine induces immunological memory in the form of memory B and T lymphocytes. Taken together, the study further supports the potential of rT2544 as a novel and improved vaccine candidate against S. Typhi.
Collapse
|