1
|
Muñoz Sandoval D, Bach FA, Ivens A, Harding AC, Smith NL, Mazurczyk M, Themistocleous Y, Edwards NJ, Silk SE, Barrett JR, Cowan GJ, Napolitani G, Savill NJ, Draper SJ, Minassian AM, Nahrendorf W, Spence PJ. Plasmodium falciparum infection induces T cell tolerance that is associated with decreased disease severity upon re-infection. J Exp Med 2025; 222:e20241667. [PMID: 40214640 PMCID: PMC11987708 DOI: 10.1084/jem.20241667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/18/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025] Open
Abstract
Immunity to severe malaria is acquired quickly, operates independently of pathogen load, and represents a highly effective form of disease tolerance. The mechanism that underpins tolerance remains unknown. We used a human rechallenge model of falciparum malaria in which healthy adult volunteers were infected three times over a 12 mo period to track the development of disease tolerance in real-time. We found that parasitemia triggered a hardwired innate immune response that led to systemic inflammation, pyrexia, and hallmark symptoms of clinical malaria across the first three infections of life. In contrast, a single infection was sufficient to reprogram T cell activation and reduce the number and diversity of effector cells upon rechallenge. Crucially, this did not silence stem-like memory cells but instead prevented the generation of cytotoxic effectors associated with autoinflammatory disease. Tolerized hosts were thus able to prevent collateral tissue damage in the absence of antiparasite immunity.
Collapse
Affiliation(s)
- Diana Muñoz Sandoval
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Instituto de Microbiologia, Universidad San Francisco de Quito, Quito, Ecuador
| | - Florian A. Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Alasdair Ivens
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Adam C. Harding
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Natasha L. Smith
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Michalina Mazurczyk
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | | | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jordan R. Barrett
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Graeme J.M. Cowan
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Giorgio Napolitani
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicholas J. Savill
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Angela M. Minassian
- The Jenner Institute, University of Oxford, Oxford, UK
- Department of Biochemistry and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Wiebke Nahrendorf
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Philip J. Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Devan J, Sandalova M, Bitterli P, Herger N, Mengis T, Brender K, Heggli I, Distler O, Dudli S. Massively parallel flow-cytometry-based screening of hematopoietic lineage cell populations from up to 25 donors simultaneously. Methods 2025; 234:45-53. [PMID: 39608688 DOI: 10.1016/j.ymeth.2024.11.014] [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: 10/10/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 11/30/2024] Open
Abstract
This study aimed to develop a method allowing high-dimensional and technically uniform screening of surface markers on cells of hematopoietic origin. High-dimensional screening of cell phenotypes is primarily the domain of single-cell RNA sequencing (RNAseq), which allows simultaneous analysis of the expression of thousands of genes in several thousands of cells. However, rare cell populations can often substantially impact tissue homeostasis or disease pathogenesis, and dysregulation of rare populations can easily be missed when only a few thousand cells are analyzed. With the presented methodological approach, it is possible to screen hundreds of markers on millions of cells in a technically uniform manner and thus identify and characterize changes in rare populations. We utilize the highly expressed markers CD45 on immune cells and CD71 on erythroid progenitors to create unique fluorescent barcodes on each of the 25 samples. Double-barcoded samples are co-stained with a broad immunophenotyping panel. The panel is designed in such a way that allows the addition of PE-labelled antibody, which was used for screening purposes. Multiplexed samples are divided into hundreds of aliquots and co-stained, each aliquot with a different PE-labelled antibody. Utilizing a broad immunophenotyping panel and machine-learning algorithms, we can predict the co-expression of hundreds of screened markers with a high degree of precision. This technique is suitable for screening immune cells in bone marrow from different locations, blood specimens, or any tissue with a substantial presence of immune cells, such as tumors or inflamed tissue areas in autoimmune conditions. It represents an approach that can significantly improve our ability to recognize dysregulated immune cell populations and, if needed, precisely target subsequent experiments covering lower cell counts such as RNAseq.
Collapse
Affiliation(s)
- Jan Devan
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland.
| | - Michaela Sandalova
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - Pamela Bitterli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - Nick Herger
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - Tamara Mengis
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - Kenta Brender
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - Irina Heggli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - Stefan Dudli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Switzerland; Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Horn V, Cancino CA, Steinheuer LM, Obermayer B, Fritz K, Nguyen AL, Juhran KS, Plattner C, Bösel D, Oldenburg L, Burns M, Schulz AR, Saliutina M, Mantzivi E, Lissner D, Conrad T, Mashreghi MF, Zundler S, Sonnenberg E, Schumann M, Haag LM, Beule D, Flatz L, Trajanoski Z, D'Haens G, Weidinger C, Mei HE, Siegmund B, Thurley K, Hegazy AN. Multimodal Profiling of Peripheral Blood Identifies Proliferating Circulating Effector CD4 + T Cells as Predictors for Response to Integrin α4β7-Blocking Therapy in Inflammatory Bowel Disease. Gastroenterology 2025; 168:327-343. [PMID: 39343250 DOI: 10.1053/j.gastro.2024.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND & AIMS Despite the success of biological therapies in treating inflammatory bowel disease, managing patients remains challenging due to the absence of reliable predictors of therapy response. METHODS In this study, we prospectively sampled 2 cohorts of patients with inflammatory bowel disease receiving the anti-integrin α4β7 antibody vedolizumab. Samples were subjected to mass cytometry; single-cell RNA sequencing; single-cell B and T cell receptor sequencing (BCR/TCR-seq); serum proteomics; and multiparametric flow cytometry to comprehensively assess vedolizumab-induced immunologic changes in the peripheral blood and their potential associations with treatment response. RESULTS Vedolizumab treatment led to substantial alterations in the abundance of circulating immune cell lineages and modified the T-cell receptor diversity of gut-homing CD4+ memory T cells. Through integration of multimodal parameters and machine learning, we identified a significant increase in proliferating CD4+ memory T cells among nonresponders before treatment compared with responders. This predictive T-cell signature demonstrated an activated T-helper 1/T-helper 17 cell phenotype and exhibited elevated levels of integrin α4β1, potentially making these cells less susceptible to direct targeting by vedolizumab. CONCLUSIONS These findings provide a reliable predictive classifier with significant implications for personalized inflammatory bowel disease management.
Collapse
Affiliation(s)
- Veronika Horn
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Camila A Cancino
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Lisa M Steinheuer
- Institute of Experimental Oncology, Biomathematics Division, University Hospital Bonn, Bonn, Germany
| | - Benedikt Obermayer
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Konstantin Fritz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Anke L Nguyen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Department of Gastroenterology, Central Clinical School, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Kim Susan Juhran
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Christina Plattner
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Diana Bösel
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Lotte Oldenburg
- Department of Gastroenterology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Marie Burns
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Axel Ronald Schulz
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Mariia Saliutina
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Eleni Mantzivi
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Donata Lissner
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Thomas Conrad
- Genomics Technology Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Core Unit Genomics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany; German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Berlin, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany
| | - Elena Sonnenberg
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Michael Schumann
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Lea-Maxie Haag
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Core Unit Bioinformatics, Berlin, Germany
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland; Department of Dermatology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Geert D'Haens
- Department of Gastroenterology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Carl Weidinger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Henrik E Mei
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany
| | - Britta Siegmund
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Kevin Thurley
- Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany; Institute of Experimental Oncology, Biomathematics Division, University Hospital Bonn, Bonn, Germany.
| | - Ahmed N Hegazy
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum, ein Institut der Leibniz-Gemeinschaft, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin Institute of Health Academy, Clinician Scientist Program, Berlin, Germany.
| |
Collapse
|
4
|
Goldner Kabeli R, Zevin S, Abargel A, Zilberberg A, Efroni S. Self-supervised learning of T cell receptor sequences exposes core properties for T cell membership. SCIENCE ADVANCES 2024; 10:eadk4670. [PMID: 38669334 PMCID: PMC11809652 DOI: 10.1126/sciadv.adk4670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 03/26/2024] [Indexed: 04/28/2024]
Abstract
The T cell receptor (TCR) repertoire is an extraordinarily diverse collection of TCRs essential for maintaining the body's homeostasis and response to threats. In this study, we compiled an extensive dataset of more than 4200 bulk TCR repertoire samples, encompassing 221,176,713 sequences, alongside 6,159,652 single-cell TCR sequences from over 400 samples. From this dataset, we then selected a representative subset of 5 million bulk sequences and 4.2 million single-cell sequences to train two specialized Transformer-based language models for bulk (CVC) and single-cell (scCVC) TCR repertoires, respectively. We show that these models successfully capture TCR core qualities, such as sharing, gene composition, and single-cell properties. These qualities are emergent in the encoded TCR latent space and enable classification into TCR-based qualities such as public sequences. These models demonstrate the potential of Transformer-based language models in TCR downstream applications.
Collapse
Affiliation(s)
- Romi Goldner Kabeli
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Avital Abargel
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Alona Zilberberg
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | | |
Collapse
|
5
|
Jiao W, Martinez M, Muntnich CB, Zuber J, Parks C, Obradovic A, Tian G, Wang Z, Long KD, Waffarn E, Frangaj K, Jones R, Gorur A, Shonts B, Rogers K, Lv G, Velasco M, Ravella S, Weiner J, Kato T, Shen Y, Fu J, Sykes M. Dynamic establishment of recipient resident memory T cell repertoire after human intestinal transplantation. EBioMedicine 2024; 101:105028. [PMID: 38422982 PMCID: PMC10944178 DOI: 10.1016/j.ebiom.2024.105028] [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: 02/07/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Understanding formation of the human tissue resident memory T cell (TRM) repertoire requires longitudinal access to human non-lymphoid tissues. METHODS By applying flow cytometry and next generation sequencing to serial blood, lymphoid tissue, and gut samples from 16 intestinal transplantation (ITx) patients, we assessed the origin, distribution, and specificity of human TRMs at phenotypic and clonal levels. FINDINGS Donor age ≥1 year and blood T cell macrochimerism (peak level ≥4%) were associated with delayed establishment of stable recipient TRM repertoires in the transplanted ileum. T cell receptor (TCR) overlap between paired gut and blood repertoires from ITx patients was significantly greater than that in healthy controls, demonstrating increased gut-blood crosstalk after ITx. Crosstalk with the circulating pool remained high for years of follow-up. TCR sequences identifiable in pre-Tx recipient gut but not those in lymphoid tissues alone were more likely to populate post-Tx ileal allografts. Clones detected in both pre-Tx gut and lymphoid tissue had distinct transcriptional profiles from those identifiable in only one tissue. Recipient T cells were distributed widely throughout the gut, including allograft and native colon, which had substantial repertoire overlap. Both alloreactive and microbe-reactive recipient T cells persisted in transplanted ileum, contributing to the TRM repertoire. INTERPRETATION Our studies reveal human intestinal TRM repertoire establishment from the circulation, preferentially involving lymphoid tissue counterparts of recipient intestinal T cell clones, including TRMs. We have described the temporal and spatial dynamics of this active crosstalk between the circulating pool and the intestinal TRM pool. FUNDING This study was funded by the National Institute of Allergy and Infectious Diseases (NIAID) P01 grant AI106697.
Collapse
Affiliation(s)
- Wenyu Jiao
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States; Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Jilin, China
| | - Mercedes Martinez
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Constanza Bay Muntnich
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Julien Zuber
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Christopher Parks
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Guangyao Tian
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Jilin, China
| | - Zicheng Wang
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, NY, United States
| | - Katherine D Long
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Elizabeth Waffarn
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Kristjana Frangaj
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Rebecca Jones
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Alaka Gorur
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Brittany Shonts
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Kortney Rogers
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Jilin, China
| | - Monica Velasco
- School of Nursing, Columbia University, New York, NY, United States
| | - Shilpa Ravella
- Department of Medicine, Columbia University, New York, NY, United States
| | - Joshua Weiner
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States; Department of Surgery, Columbia University, New York, NY, United States
| | - Tomoaki Kato
- Department of Surgery, Columbia University, New York, NY, United States
| | - Yufeng Shen
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, NY, United States
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States.
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, United States; Department of Surgery, Columbia University, New York, NY, United States; Department of Microbiology & Immunology, Columbia University, New York, NY, United States.
| |
Collapse
|
6
|
Martin LB, Khanam F, Qadri F, Khalil I, Sikorski MJ, Baker S. Vaccine value profile for Salmonella enterica serovar Paratyphi A. Vaccine 2023; 41 Suppl 2:S114-S133. [PMID: 37951691 DOI: 10.1016/j.vaccine.2023.01.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/27/2022] [Accepted: 01/23/2023] [Indexed: 11/14/2023]
Abstract
In Asia, there are an estimated 12 million annual cases of enteric fever, a potentially fatal systemic bacterial infection caused by Salmonella enterica serovars Typhi (STy) and Paratyphi A (SPA). The recent availability of typhoid conjugate vaccines (TCV), an increasing incidence of disease caused by SPA and growing antimicrobial resistance (AMR) across the genus Salmonella makes a bivalent STy/SPA vaccine a useful public health proposition. The uptake of a stand-alone paratyphoid vaccine is likely low thus, there is a pipeline of bivalent STy/SPA candidate vaccines. Several candidates are close to entering clinical trials, which if successful should facilitate a more comprehensive approach for enteric fever control. Additionally, the World Health Organization (WHO) has made advancing the development of vaccines that protect young children and working aged adults against both agents of enteric fever a priority objective. This "Vaccine Value Profile" (VVP) addresses information related predominantly to invasive disease caused by SPA prevalent in Asia. Information is included on stand-alone SPA candidate vaccines and candidate vaccines targeting SPA combined with STy. Out of scope for the first version of this VVP is a wider discussion on the development of a universal Salmonella combination candidate vaccine, addressing both enteric fever and invasive non-typhoidal Salmonella disease, for use globally. This VVP is a detailed, high-level assessment of existing, publicly available information to inform and contextualize the public health, economic, and societal potential of pipeline vaccines and vaccine-like products for SPA. Future versions of this VVP will be updated to reflect ongoing activities such as vaccine development strategies and "Full Vaccine Value Assessment" that will inform the value proposition of an SPA vaccine. This VVP was developed by an expert working group from academia, non-profit organizations, public-private partnerships, and multi-lateral organizations as well as in collaboration with stakeholders from the WHO South-East Asian Region. All contributors have extensive expertise on various elements of the VVP for SPA and collectively aimed to identify current research and knowledge gaps.
Collapse
Affiliation(s)
- Laura B Martin
- Independent Consultant (current affiliation US Pharmacopeia Convention), USA.
| | - Farhana Khanam
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Bangladesh.
| | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Bangladesh.
| | | | | | - Stephen Baker
- University of Cambridge School of Clinical Medicine, UK.
| |
Collapse
|
7
|
Bach FA, Muñoz Sandoval D, Mazurczyk M, Themistocleous Y, Rawlinson TA, Harding AC, Kemp A, Silk SE, Barrett JR, Edwards NJ, Ivens A, Rayner JC, Minassian AM, Napolitani G, Draper SJ, Spence PJ. A systematic analysis of the human immune response to Plasmodium vivax. J Clin Invest 2023; 133:e152463. [PMID: 37616070 PMCID: PMC10575735 DOI: 10.1172/jci152463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/22/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUNDThe biology of Plasmodium vivax is markedly different from that of P. falciparum; how this shapes the immune response to infection remains unclear. To address this shortfall, we inoculated human volunteers with a clonal field isolate of P. vivax and tracked their response through infection and convalescence.METHODSParticipants were injected intravenously with blood-stage parasites and infection dynamics were tracked in real time by quantitative PCR. Whole blood samples were used for high dimensional protein analysis, RNA sequencing, and cytometry by time of flight, and temporal changes in the host response to P. vivax were quantified by linear regression. Comparative analyses with P. falciparum were then undertaken using analogous data sets derived from prior controlled human malaria infection studies.RESULTSP. vivax rapidly induced a type I inflammatory response that coincided with hallmark features of clinical malaria. This acute-phase response shared remarkable overlap with that induced by P. falciparum but was significantly elevated (at RNA and protein levels), leading to an increased incidence of pyrexia. In contrast, T cell activation and terminal differentiation were significantly increased in volunteers infected with P. falciparum. Heterogeneous CD4+ T cells were found to dominate this adaptive response and phenotypic analysis revealed unexpected features normally associated with cytotoxicity and autoinflammatory disease.CONCLUSIONP. vivax triggers increased systemic interferon signaling (cf P. falciparum), which likely explains its reduced pyrogenic threshold. In contrast, P. falciparum drives T cell activation far in excess of P. vivax, which may partially explain why falciparum malaria more frequently causes severe disease.TRIAL REGISTRATIONClinicalTrials.gov NCT03797989.FUNDINGThe European Union's Horizon 2020 Research and Innovation programme, the Wellcome Trust, and the Royal Society.
Collapse
Affiliation(s)
- Florian A. Bach
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Diana Muñoz Sandoval
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
- Insitute of Microbiology, Universidad San Francisco de Quito, Quito, Ecuador
| | | | | | | | - Adam C. Harding
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Alison Kemp
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Jordan R. Barrett
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Alasdair Ivens
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Angela M. Minassian
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Giorgio Napolitani
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, and
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Philip J. Spence
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
8
|
Ma R, Rei M, Woodhouse I, Ferris K, Kirschner S, Chandran A, Gileadi U, Chen JL, Pereira Pinho M, Ariosa-Morejon Y, Kriaucionis S, Ternette N, Koohy H, Ansorge O, Ogg G, Plaha P, Cerundolo V. Decitabine increases neoantigen and cancer testis antigen expression to enhance T-cell-mediated toxicity against glioblastoma. Neuro Oncol 2022; 24:2093-2106. [PMID: 35468205 PMCID: PMC9713507 DOI: 10.1093/neuonc/noac107] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults. Despite maximal treatment, median survival remains dismal at 14-24 months. Immunotherapies, such as checkpoint inhibition, have revolutionized management of some cancers but have little benefit for GBM patients. This is, in part, due to the low mutational and neoantigen burden in this immunogenically "cold" tumor. METHODS U87MG and patient-derived cell lines were treated with 5-aza-2'-deoxycytidine (DAC) and underwent whole-exome and transcriptome sequencing. Cell lines were then subjected to cellular assays with neoantigen and cancer testis antigen (CTA) specific T cells. RESULTS We demonstrate that DAC increases neoantigen and CTA mRNA expression through DNA hypomethylation. This results in increased neoantigen presentation by MHC class I in tumor cells, leading to increased neoantigen- and CTA-specific T-cell activation and killing of DAC-treated cancer cells. In addition, we show that patients have endogenous cancer-specific T cells in both tumor and blood, which show increased tumor-specific activation in the presence of DAC-treated cells. CONCLUSIONS Our work shows that DAC increases GBM immunogenicity and consequent susceptibility to T-cell responses in vitro. Our results support a potential use of DAC as a sensitizing agent for immunotherapy.
Collapse
Affiliation(s)
- Ruichong Ma
- Corresponding Authors: Ruichong Ma, DPhil, Department of neurosurgery, Level 3 West wing, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK ()
| | - Margarida Rei
- Margarida Rei, PhD, Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK ()
| | - Isaac Woodhouse
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Centre for Cellular and Medical Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katherine Ferris
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Sophie Kirschner
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Anandhakumar Chandran
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ji-Li Chen
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Mariana Pereira Pinho
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Yoanna Ariosa-Morejon
- Centre for Cellular and Medical Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Skirmantas Kriaucionis
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nicola Ternette
- Centre for Cellular and Medical Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The Jenner Institute, University of Oxford, Oxford, UK (Y.A-M., N.T.)
| | - Hashem Koohy
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University ofOxford, UK
| | - Graham Ogg
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Puneet Plaha
- Department of Neurosurgery, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University ofOxford, UK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Sztein MB, Booth JS. Controlled human infectious models, a path forward in uncovering immunological correlates of protection: Lessons from enteric fevers studies. Front Microbiol 2022; 13:983403. [PMID: 36204615 PMCID: PMC9530043 DOI: 10.3389/fmicb.2022.983403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Enteric infectious diseases account for more than a billion disease episodes yearly worldwide resulting in approximately 2 million deaths, with children under 5 years old and the elderly being disproportionally affected. Enteric pathogens comprise viruses, parasites, and bacteria; the latter including pathogens such as Salmonella [typhoidal (TS) and non-typhoidal (nTS)], cholera, Shigella and multiple pathotypes of Escherichia coli (E. coli). In addition, multi-drug resistant and extensively drug-resistant (XDR) strains (e.g., S. Typhi H58 strain) of enteric bacteria are emerging; thus, renewed efforts to tackle enteric diseases are required. Many of these entero-pathogens could be controlled by oral or parenteral vaccines; however, development of new, effective vaccines has been hampered by lack of known immunological correlates of protection (CoP) and limited knowledge of the factors contributing to protective responses. To fully comprehend the human response to enteric infections, an invaluable tool that has recently re-emerged is the use of controlled human infection models (CHIMs) in which participants are challenged with virulent wild-type (wt) organisms. CHIMs have the potential to uncover immune mechanisms and identify CoP to enteric pathogens, as well as to evaluate the efficacy of therapeutics and vaccines in humans. CHIMs have been used to provide invaluable insights in the pathogenesis, host-pathogen interaction and evaluation of vaccines. Recently, several Oxford typhoid CHIM studies have been performed to assess the role of multiple cell types (B cells, CD8+ T, Tregs, MAIT, Monocytes and DC) during S. Typhi infection. One of the key messages that emerged from these studies is that baseline antigen-specific responses are important in that they can correlate with clinical outcomes. Additionally, volunteers who develop typhoid disease (TD) exhibit higher levels and more activated cell types (e.g., DC and monocytes) which are nevertheless defective in discrete signaling pathways. Future critical aspects of this research will involve the study of immune responses to enteric infections at the site of entry, i.e., the intestinal mucosa. This review will describe our current knowledge of immunity to enteric fevers caused byS. Typhi and S. Paratyphi A, with emphasis on the contributions of CHIMs to uncover the complex immunological responses to these organisms and provide insights into the determinants of protective immunity.
Collapse
Affiliation(s)
- Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: Marcelo B. Sztein,
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Jayaum S. Booth,
| |
Collapse
|
10
|
Slack E, Diard M. Resistance is futile? Mucosal immune mechanisms in the context of microbial ecology and evolution. Mucosal Immunol 2022; 15:1188-1198. [PMID: 36329192 PMCID: PMC9705250 DOI: 10.1038/s41385-022-00574-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
In the beginning it was simple: we injected a protein antigen and studied the immune responses against the purified protein. This elegant toolbox uncovered thousands of mechanisms via which immune cells are activated. However, when we consider immune responses against real infectious threats, this elegant simplification misses half of the story: the infectious agents are typically evolving orders-of-magnitude faster than we are. Nowhere is this more pronounced than in the mammalian large intestine. A bacterium representing only 0.1% of the human gut microbiota will have a population size of 109 clones, each actively replicating. Moreover, the evolutionary pressure from other microbes is at least as profound as direct effects of the immune system. Therefore, to really understand intestinal immune mechanisms, we need to understand both the host response and how rapid microbial evolution alters the apparent outcome of the response. In this review we use the examples of intestinal inflammation and secretory immunoglobulin A (SIgA) to highlight what is already known (Fig. 1). Further, we will explore how these interactions can inform immunotherapy and prophylaxis. This has major implications for how we design effective mucosal vaccines against increasingly drug-resistant bacterial pathogens Fig. 1 THE IMMUNE RESPONSE SHAPES THE FITNESS LANDSCAPE IN THE GASTRO-INTESTINAL TRACT.: The red arrows depict possible evolutionary paths of a novel colonizer along adaptive peaks in the intestinal fitness landscapes that change with the status of the host immune system. The flat surfaces represent the non-null fitness baselines (values x or y) at which a bacterium can establish at minimum carrying capacity. a In the healthy gut, metabolic competence, resistance to aggressions by competitors and predators, swift adaptation to rapid fluctuations as well as surviving acidic pH and the flow of the intestinal content, represent potent selective pressures and as many opportunities for bacteria to increase fitness by phenotypic or genetic variations. b When pathogens trigger acute inflammation, bacteria must adapt to iron starvation, killing by immune cells and antimicrobial peptides, and oxidative stress, while new metabolic opportunities emerge. c When high-affinity SIgA are produced against a bacterium, e.g., after oral vaccination, escape of SIgA by altering or losing surface epitopes becomes crucial for maximum fitness. However, escaping polyvalent SIgA responses after vaccination with "evolutionary trap" vaccines leads to evolutionary trade-offs: A fitness maximum is reached in the vaccinated host gut that represents a major disadvantage for transmission into naïve hosts (fitness diminished below x) (d).
Collapse
Affiliation(s)
- Emma Slack
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland.
- Botnar Research Institute for Child Health, Basel, Switzerland.
| | - Médéric Diard
- Botnar Research Institute for Child Health, Basel, Switzerland.
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Cassotta A, Goldstein JD, Durini G, Jarrossay D, Baggi Menozzi F, Venditti M, Russo A, Falcone M, Lanzavecchia A, Gagliardi MC, Latorre D, Sallusto F. Broadly reactive human CD4 + T cells against Enterobacteriaceae are found in the naïve repertoire and are clonally expanded in the memory repertoire. Eur J Immunol 2021; 51:648-661. [PMID: 33226131 PMCID: PMC7986685 DOI: 10.1002/eji.202048630] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 09/15/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022]
Abstract
Enterobacteriaceae are a large family of Gram-negative bacteria that includes both commensals and opportunistic pathogens. The latter can cause severe nosocomial infections, with outbreaks of multi-antibiotics resistant strains, thus being a major public health threat. In this study, we report that Enterobacteriaceae-reactive memory Th cells were highly enriched in a CCR6+ CXCR3+ Th1*/17 cell subset and produced IFN-γ, IL-17A, and IL-22. This T cell subset was severely reduced in septic patients with K. pneumoniae bloodstream infection who also selectively lacked circulating K. pneumonie-reactive T cells. By combining heterologous antigenic stimulation, single cell cloning and TCR Vβ sequencing, we demonstrate that a large fraction of memory Th cell clones was broadly cross-reactive to several Enterobacteriaceae species. These cross-reactive Th cell clones were expanded in vivo and a large fraction of them recognized the conserved outer membrane protein A antigen. Interestingly, Enterobacteriaceae broadly cross-reactive T cells were also prominent among in vitro primed naïve T cells. Collectively, these data point to the existence of immunodominant T cell epitopes shared among different Enterobacteriaceae species and targeted by cross-reactive T cells that are readily found in the pre-immune repertoire and are clonally expanded in the memory repertoire.
Collapse
Affiliation(s)
- Antonino Cassotta
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
- Institute of MicrobiologyETH ZurichSwitzerland
| | - Jérémie D. Goldstein
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
| | - Greta Durini
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
| | - David Jarrossay
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
| | | | - Mario Venditti
- Department of Public Health and Infectious DiseasesSapienza University of RomeRomeItaly
| | - Alessandro Russo
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Marco Falcone
- Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Antonio Lanzavecchia
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
| | | | - Daniela Latorre
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
- Institute of MicrobiologyETH ZurichSwitzerland
| | - Federica Sallusto
- Institute for Research in BiomedicineUniversità della Svizzera italianaBellinzonaSwitzerland
- Institute of MicrobiologyETH ZurichSwitzerland
| |
Collapse
|
12
|
Moody DB. Remembering Enzo Cerundolo. Mol Immunol 2020; 129:53-55. [PMID: 33309954 DOI: 10.1016/j.molimm.2020.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 11/27/2022]
Affiliation(s)
- D Branch Moody
- Brigham and Women's Hospital, Division of Rheumatology, Inflammation and Immunity, Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, United States.
| |
Collapse
|
13
|
Gibani MM, Jin C, Shrestha S, Moore M, Norman L, Voysey M, Jones E, Blackwell L, Thomaides-Brears H, Hill J, Blohmke CJ, Dobinson HC, Baker P, Jones C, Campbell D, Mujadidi YF, Plested E, Preciado-Llanes L, Napolitani G, Simmons A, Gordon MA, Angus B, Darton TC, Cerundulo V, Pollard AJ. Homologous and heterologous re-challenge with Salmonella Typhi and Salmonella Paratyphi A in a randomised controlled human infection model. PLoS Negl Trop Dis 2020; 14:e0008783. [PMID: 33079959 PMCID: PMC7598925 DOI: 10.1371/journal.pntd.0008783] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/30/2020] [Accepted: 09/08/2020] [Indexed: 11/19/2022] Open
Abstract
Enteric fever is a systemic infection caused by Salmonella Typhi or Paratyphi A. In many endemic areas, these serovars co-circulate and can cause multiple infection-episodes in childhood. Prior exposure is thought to confer partial, but incomplete, protection against subsequent attacks of enteric fever. Empirical data to support this hypothesis are limited, and there are few studies describing the occurrence of heterologous-protection between these closely related serovars. We performed a challenge-re-challenge study using a controlled human infection model (CHIM) to investigate the extent of infection-derived immunity to Salmonella Typhi or Paratyphi A infection. We recruited healthy volunteers into two groups: naïve volunteers with no prior exposure to Salmonella Typhi/Paratyphi A and volunteers previously-exposed to Salmonella Typhi or Paratyphi A in earlier CHIM studies. Within each group, participants were randomised 1:1 to oral challenge with either Salmonella Typhi (104 CFU) or Paratyphi A (103 CFU). The primary objective was to compare the attack rate between naïve and previously challenged individuals, defined as the proportion of participants per group meeting the diagnostic criteria of temperature of ≥38°C persisting for ≥12 hours and/or S. Typhi/Paratyphi bacteraemia up to day 14 post challenge. The attack-rate in participants who underwent homologous re-challenge with Salmonella Typhi was reduced compared with challenged naïve controls, although this reduction was not statistically significant (12/27[44%] vs. 12/19[63%]; Relative risk 0.70; 95% CI 0.41-1.21; p = 0.24). Homologous re-challenge with Salmonella Paratyphi A also resulted in a lower attack-rate than was seen in challenged naïve controls (3/12[25%] vs. 10/18[56%]; RR0.45; 95% CI 0.16-1.30; p = 0.14). Evidence of protection was supported by a post hoc analysis in which previous exposure was associated with an approximately 36% and 57% reduced risk of typhoid or paratyphoid disease respectively on re-challenge. Individuals who did not develop enteric fever on primary exposure were significantly more likely to be protected on re-challenge, compared with individuals who developed disease on primary exposure. Heterologous re-challenge with Salmonella Typhi or Salmonella Paratyphi A was not associated with a reduced attack rate following challenge. Within the context of the model, prior exposure was not associated with reduced disease severity, altered microbiological profile or boosting of humoral immune responses. We conclude that prior Salmonella Typhi and Paratyphi A exposure may confer partial but incomplete protection against subsequent infection, but with a comparable clinical and microbiological phenotype. There is no demonstrable cross-protection between these serovars, consistent with the co-circulation of Salmonella Typhi and Paratyphi A. Collectively, these data are consistent with surveillance and modelling studies that indicate multiple infections can occur in high transmission settings, supporting the need for vaccines to reduce the burden of disease in childhood and achieve disease control. Trial registration NCT02192008; clinicaltrials.gov.
Collapse
Affiliation(s)
- Malick M. Gibani
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
- Department of Infectious Diseases, Imperial College London, United Kingdom
| | - Celina Jin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Sonu Shrestha
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Maria Moore
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Lily Norman
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Elizabeth Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Luke Blackwell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Helena Thomaides-Brears
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Christoph J. Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Hazel C. Dobinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Philip Baker
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Danielle Campbell
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Yama F. Mujadidi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Lorena Preciado-Llanes
- Institute for Infection and Global Health, University of Liverpool, United Kingdom
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Giorgio Napolitani
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Alison Simmons
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Melita A. Gordon
- Institute for Infection and Global Health, University of Liverpool, United Kingdom
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Vincenzo Cerundulo
- Medical Research Council Human Immunology Unit, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, United Kingdom
| |
Collapse
|
14
|
Zhang T, Warden AR, Li Y, Ding X. Progress and applications of mass cytometry in sketching immune landscapes. Clin Transl Med 2020; 10:e206. [PMID: 33135337 PMCID: PMC7556381 DOI: 10.1002/ctm2.206] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Recently emerged mass cytometry (cytometry by time-of-flight [CyTOF]) technology permits the identification and quantification of inherently diverse cellular systems, and the simultaneous measurement of functional attributes at the single-cell resolution. By virtue of its multiplex ability with limited need for compensation, CyTOF has led a critical role in immunological research fields. Here, we present an overview of CyTOF, including the introduction of CyTOF principle and advantages that make it a standalone tool in deciphering immune mysteries. We then discuss the functional assays, introduce the bioinformatics to interpret the data yield via CyTOF, and depict the emerging clinical and research applications of CyTOF technology in sketching immune landscape in a wide variety of diseases.
Collapse
Affiliation(s)
- Ting Zhang
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Antony R. Warden
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yiyang Li
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Xianting Ding
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
15
|
Rapaka RR, Wahid R, Fresnay S, Booth JS, Darton TC, Jones C, Waddington CS, Levine MM, Pollard AJ, Sztein MB. Human Salmonella Typhi exposure generates differential multifunctional cross-reactive T-cell memory responses against Salmonella Paratyphi and invasive nontyphoidal Salmonella. Clin Transl Immunology 2020; 9:e1178. [PMID: 33005416 PMCID: PMC7512505 DOI: 10.1002/cti2.1178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Objective There are no vaccines for most of the major invasive Salmonella strains causing severe infection in humans. We evaluated the specificity of adaptive T memory cell responses generated after Salmonella Typhi exposure in humans against other major invasive Salmonella strains sharing capacity for dissemination. Methods T memory cells from eleven volunteers who underwent controlled oral challenge with wtS. Typhi were characterised by flow cytometry for cross‐reactive cellular cytokine/chemokine effector responses or evidence of degranulation upon stimulation with autologous B‐lymphoblastoid cells infected with either S. Typhi, Salmonella Paratyphi A (PA), S. Paratyphi B (PB) or an invasive nontyphoidal Salmonella strain of the S. Typhimurium serovar (iNTSTy). Results Blood T‐cell effector memory (TEM) responses after exposure to S. Typhi in humans evolve late, peaking weeks after infection in most volunteers. Induced multifunctional CD4+ Th1 and CD8+ TEM cells elicited after S. Typhi challenge were cross‐reactive with PA, PB and iNTSTy. The magnitude of multifunctional CD4+ TEM cell responses to S. Typhi correlated with induction of cross‐reactive multifunctional CD8+ TEM cells against PA, PB and iNTSTy. Highly multifunctional subsets and T central memory and T effector memory cells that re‐express CD45 (TEMRA) demonstrated less heterologous T‐cell cross‐reactivity, and multifunctional Th17 elicited after S. Typhi challenge was not cross‐reactive against other invasive Salmonella. Conclusion Gaps in cross‐reactive immune effector functions in human T‐cell memory compartments were highly dependent on invasive Salmonella strain, underscoring the importance of strain‐dependent vaccination in the design of T‐cell‐based vaccines for invasive Salmonella.
Collapse
Affiliation(s)
- Rekha R Rapaka
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Rezwanul Wahid
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Stephanie Fresnay
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA.,Present address: Stephanie Fresnay GlaxoSmithKline Rockville MD USA
| | - Jayaum S Booth
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Thomas C Darton
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK.,Present address: Thomas C Darton University of Sheffield Medical School Sheffield UK
| | - Claire Jones
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK
| | - Claire S Waddington
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK.,Present address: University of Cambridge Cambridge UK
| | - Myron M Levine
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Andrew J Pollard
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| |
Collapse
|
16
|
Evasion of MAIT cell recognition by the African Salmonella Typhimurium ST313 pathovar that causes invasive disease. Proc Natl Acad Sci U S A 2020; 117:20717-20728. [PMID: 32788367 PMCID: PMC7456131 DOI: 10.1073/pnas.2007472117] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are innate T lymphocytes activated by bacteria that produce vitamin B2 metabolites. Mouse models of infection have demonstrated a role for MAIT cells in antimicrobial defense. However, proposed protective roles of MAIT cells in human infections remain unproven and clinical conditions associated with selective absence of MAIT cells have not been identified. We report that typhoidal and nontyphoidal Salmonella enterica strains activate MAIT cells. However, S. Typhimurium sequence type 313 (ST313) lineage 2 strains, which are responsible for the burden of multidrug-resistant nontyphoidal invasive disease in Africa, escape MAIT cell recognition through overexpression of ribB This bacterial gene encodes the 4-dihydroxy-2-butanone-4-phosphate synthase enzyme of the riboflavin biosynthetic pathway. The MAIT cell-specific phenotype did not extend to other innate lymphocytes. We propose that ribB overexpression is an evolved trait that facilitates evasion from immune recognition by MAIT cells and contributes to the invasive pathogenesis of S. Typhimurium ST313 lineage 2.
Collapse
|
17
|
Booth JS, Goldberg E, Patil SA, Barnes RS, Greenwald BD, Sztein MB. Effect of the live oral attenuated typhoid vaccine, Ty21a, on systemic and terminal ileum mucosal CD4+ T memory responses in humans. Int Immunol 2020; 31:101-116. [PMID: 30346608 PMCID: PMC6376105 DOI: 10.1093/intimm/dxy070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/07/2018] [Indexed: 01/01/2023] Open
Abstract
Our current understanding of CD4+ T-cell-mediated immunity (CMI) elicited by the oral live attenuated typhoid vaccine Ty21a is primarily derived from studies using peripheral blood. Very limited data are available in humans regarding mucosal immunity (especially CD4+ T) at the site of infection (e.g. terminal ileum; TI). Here using multiparametric flow cytometry, we examined the effect of Ty21a immunization on TI-lamina propria mononuclear cells (LPMC) and peripheral blood CD4+ T memory (TM) subsets in volunteers undergoing routine colonoscopy. Interestingly, we observed significant increases in the frequencies of LPMC CD4+ T cells following Ty21a immunization, restricted to the T effector/memory (TEM)-CD45RA+ (TEMRA) subset. Importantly, Ty21a immunization elicited Salmonella Typhi-responsive LPMC CD4+ T cells in all major TM subsets [interferon (IFN)γ and interleukin (IL)-17A in TEM; IFNγ and macrophage inflammatory protein (MIP)1β in T central/memory (TCM); and IL-2 in TEMRA]. Subsequently, we analyzed LPMC S. Typhi-responsive CD4+ T cells in depth for multifunctional (MF) effectors. We found that LPMC CD4+ TEM responses were mostly MF, except for those cells exhibiting the characteristics associated with IL-17A responses. Finally, we compared mucosal to systemic responses and observed that LPMC CD4+S. Typhi-specific responses were unique and distinct from their systemic counterparts. This study provides the first demonstration of S. Typhi-specific CD4+ TM responses in the human TI mucosa and provides valuable information about the generation of mucosal immune responses following oral Ty21a immunization.
Collapse
Affiliation(s)
- Jayaum S Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eric Goldberg
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Seema A Patil
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robin S Barnes
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bruce D Greenwald
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.,Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Whole blood preservation methods alter chemokine receptor detection in mass cytometry experiments. J Immunol Methods 2020; 476:112673. [DOI: 10.1016/j.jim.2019.112673] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 12/21/2022]
|
19
|
Chng MHY, Lim MQ, Rouers A, Becht E, Lee B, MacAry PA, Lye DC, Leo YS, Chen J, Fink K, Rivino L, Newell EW. Large-Scale HLA Tetramer Tracking of T Cells during Dengue Infection Reveals Broad Acute Activation and Differentiation into Two Memory Cell Fates. Immunity 2019; 51:1119-1135.e5. [PMID: 31757672 DOI: 10.1016/j.immuni.2019.10.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 08/01/2019] [Accepted: 10/21/2019] [Indexed: 12/25/2022]
Abstract
T cells play important multifaceted roles during dengue infection, and understanding their responses is important for defining correlates of protective immunity and identifying effective vaccine antigens. Using mass cytometry and a highly multiplexed peptide-HLA (human leukocyte antigen) tetramer staining strategy, we probed T cells from dengue patients-a total of 430 dengue and control candidate epitopes-together with key markers of activation, trafficking, and differentiation. During acute disease, dengue-specific CD8+ T cells expressed a distinct profile of activation and trafficking receptors that distinguished them from non-dengue-specific T cells. During convalescence, dengue-specific T cells differentiated into two major cell fates, CD57+ CD127--resembling terminally differentiated senescent memory cells and CD127+ CD57--resembling proliferation-capable memory cells. Validation in an independent cohort showed that these subsets remained at elevated frequencies up to one year after infection. These analyses aid our understanding of the generation of T cell memory in dengue infection or vaccination.
Collapse
Affiliation(s)
- Melissa Hui Yen Chng
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Mei Qiu Lim
- Emerging Infectious Diseases Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Angeline Rouers
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Etienne Becht
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Bernett Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Paul A MacAry
- Immunology Programme, Department of Microbiology and Immunology, Life Science Institute, National University of Singapore, Singapore 117456, Singapore
| | - David Chien Lye
- National Centre for Infectious Diseases, Singapore 308442, Singapore; Tan Tock Seng Hospital, Singapore 308433, Singapore; Lee Kong Chian School of Medicine, Singapore 308232, Singapore; Yong Loo Lin School of Medicine, Singapore 119228, Singapore
| | - Yee Sin Leo
- National Centre for Infectious Diseases, Singapore 308442, Singapore; Tan Tock Seng Hospital, Singapore 308433, Singapore; Lee Kong Chian School of Medicine, Singapore 308232, Singapore; Yong Loo Lin School of Medicine, Singapore 119228, Singapore; Saw Swee Hock School of Public Health, Singapore 117549, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Katja Fink
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
| | - Laura Rivino
- Emerging Infectious Diseases Programme, Duke-NUS Medical School, Singapore 169857, Singapore; School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK.
| | - Evan W Newell
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
20
|
Zvyagin IV, Tsvetkov VO, Chudakov DM, Shugay M. An overview of immunoinformatics approaches and databases linking T cell receptor repertoires to their antigen specificity. Immunogenetics 2019; 72:77-84. [PMID: 31741011 DOI: 10.1007/s00251-019-01139-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/16/2019] [Indexed: 11/26/2022]
Abstract
Recent advances in molecular and bioinformatic methods have greatly improved our ability to study the formation of an adaptive immune response towards foreign pathogens, self-antigens, and cancer neoantigens. T cell receptors (TCR) are the key players in this process that recognize peptides presented by major histocompatibility complex (MHC). Owing to the huge diversity of both TCR sequence variants and peptides they recognize, accumulation and complex analysis of large amounts of TCR-antigen specificity data is required for understanding the structure and features of adaptive immune responses towards pathogens, vaccines, cancer, as well as autoimmune responses. In the present review, we summarize recent efforts on gathering and interpreting TCR-antigen specificity data and outline the critical role of tighter integration with other immunoinformatics data sources that include epitope MHC restriction, TCR repertoire structure models, and TCR/peptide/MHC structural data. We suggest that such integration can lead to the ability to accurately annotate individual TCR repertoires, efficiently estimate epitope and neoantigen immunogenicity, and ultimately, in silico identify TCRs specific to yet unstudied antigens and predict self-peptides related to autoimmunity.
Collapse
Affiliation(s)
- Ivan V Zvyagin
- Pirogov Russian Medical State University, Moscow, Russia
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Vasily O Tsvetkov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Dmitry M Chudakov
- Pirogov Russian Medical State University, Moscow, Russia
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Mikhail Shugay
- Pirogov Russian Medical State University, Moscow, Russia.
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia.
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.
| |
Collapse
|
21
|
Salerno-Gonçalves R, Tettelin H, Luo D, Guo Q, Ardito MT, Martin WD, De Groot AS, Sztein MB. Differential functional patterns of memory CD4 + and CD8 + T-cells from volunteers immunized with Ty21a typhoid vaccine observed using a recombinant Escherichia coli system expressing S. Typhi proteins. Vaccine 2019; 38:258-270. [PMID: 31629569 DOI: 10.1016/j.vaccine.2019.10.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 02/01/2023]
Abstract
It is widely accepted that CD4+ and CD8+ T-cells play a significant role in protection against Salmonella enterica serovar Typhi (S. Typhi), the causative agent of the typhoid fever. However, the antigen specificity of these T-cells remains largely unknown. Previously, we demonstrated the feasibility of using a recombinant Escherichia coli (E. coli) expression system to uncover the antigen specificity of CD4+ and CD8+ T cells. Here, we expanded these studies to include the evaluation of 12 additional S. Typhi proteins: 4 outer membrane proteins (OmpH, OmpL, OmpR, OmpX), 3 Vi-polysaccharide biosynthesis proteins (TviA, TviB, TviE), 3 cold shock proteins (CspA, CspB, CspC), and 2 conserved hypothetical proteins (Chp 1 and Chp2), all selected based on the bioinformatic analyses of the content of putative T-cell epitopes. CD4+ and CD8+ T cells from 15 adult volunteers, obtained before and 42 days after immunization with oral live attenuated Ty21a vaccine, were assessed for their functionality (i.e., production of cytokines and cytotoxic expression markers in response to stimulation with selected antigens) as measured by flow cytometry. Although volunteers differed on their T-cell antigen specificity, we observed T-cell immune responses against all S. Typhi proteins evaluated. These responses included 9 proteins, OmpH, OmpR, TviA, TviE, CspA, CspB, CspC, Chp 1 and Chp 2, which have not been previously reported to elicit T-cell responses. Interestingly, we also observed that, regardless of the protein, the functional patterns of the memory T-cells were different between CD4+ and CD8+ T cells. In sum, these studies demonstrated the feasibility of using bioinformatic analysis and the E. coli expressing system described here to uncover novel immunogenic T-cell proteins that could serve as potential targets for the production of protein-based vaccines.
Collapse
Affiliation(s)
- Rosângela Salerno-Gonçalves
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA.
| | - Hervé Tettelin
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, 670 West Baltimore Street, HSF3, Baltimore, MD 21201, USA
| | - David Luo
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA
| | - Qin Guo
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, 670 West Baltimore Street, HSF3, Baltimore, MD 21201, USA
| | - Matthew T Ardito
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - William D Martin
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - Anne S De Groot
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA
| |
Collapse
|
22
|
Wang N, Knodler LA, Strugnell RA. Typhoid Fever: The More We Learn, the Less We Know (Apologies, Albert Einstein). Cell Host Microbe 2019; 26:303-306. [PMID: 31513767 DOI: 10.1016/j.chom.2019.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In this issue of Cell Host & Microbe, Karlinsey et al. (2019) combine TraDIS with humanized mice to identify genes required for early replication of Salmonella Typhi in vivo. Surprisingly, some expected virulence traits and genes appear dispensable in the replication of S. Typhi, supporting findings from a recent human challenge study by Gibani et al. (2019).
Collapse
Affiliation(s)
- Nancy Wang
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Leigh A Knodler
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Washington State University Paul G. Allen School for Global Animal Health, Pullman, WA, USA
| | - Richard A Strugnell
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
23
|
Parween F, Yadav J, Qadri A. The Virulence Polysaccharide of Salmonella Typhi Suppresses Activation of Rho Family GTPases to Limit Inflammatory Responses From Epithelial Cells. Front Cell Infect Microbiol 2019; 9:141. [PMID: 31134159 PMCID: PMC6517557 DOI: 10.3389/fcimb.2019.00141] [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: 01/30/2019] [Accepted: 04/16/2019] [Indexed: 12/20/2022] Open
Abstract
Vi capsular polysaccharide (Vi) is a major virulence factor of human typhoid-causing pathogen Salmonella enterica serovar Typhi (S. Typhi). It distinguishes S. Typhi from closely related non-typhoidal Salmonella serovars such as S. Typhimurium which do not normally cause systemic infection in humans. Vi not only forms a capsule around S. Typhi but it is also readily released from this pathogen. We have previously reported that Vi targets prohibitin to inhibit cellular responses activated through immune receptors. Here, we show that engagement of membrane prohibitin with Vi prevents Salmonella-induced activation of small Rho-family GTPases, Rac1, and Cdc42, and suppresses actin cytoskeletal rearrangements resulting in reduced invasion and highly subdued inflammatory responses. Cells infected with S. Typhimurium in the presence of Vi show poor activation of NF-kB and MAP-kinase pathways of intracellular signaling. Treatment with Vi brings about redistribution of Rac-1, prohibitin, and ganglioside GM1 in membrane raft domains. Vi-mediated interference with activation of Rho-family GTPases represents a previously unrecognized mechanism by which S. Typhi can limit its invasion and alarming of the host.
Collapse
Affiliation(s)
- Farhat Parween
- Hybridoma Laboratory, National Institute of Immunology, New Delhi, India
| | - Jitender Yadav
- Hybridoma Laboratory, National Institute of Immunology, New Delhi, India
| | - Ayub Qadri
- Hybridoma Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
24
|
Yeung A, Hale C, Clare S, Palmer S, Bartholdson Scott J, Baker S, Dougan G. Using a Systems Biology Approach To Study Host-Pathogen Interactions. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0021-2019. [PMID: 30953425 PMCID: PMC11590422 DOI: 10.1128/microbiolspec.bai-0021-2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Indexed: 12/18/2022] Open
Abstract
The rapid development of genomics and other "-omics" approaches has significantly impacted how we have investigated host-pathogen interactions since the turn of the millennium. Technologies such as next-generation sequencing, stem cell biology, and high-throughput proteomics have transformed the scale and sensitivity with which we interrogate biological samples. These approaches are impacting experimental design in the laboratory and transforming clinical management in health care systems. Here, we review this area from the perspective of research on bacterial pathogens.
Collapse
Affiliation(s)
- Amy Yeung
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Christine Hale
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Simon Clare
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Sophie Palmer
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Josefin Bartholdson Scott
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Stephen Baker
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
- Oxford University Clinical Research Unit, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Gordon Dougan
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
25
|
Aulicino A, Rue-Albrecht KC, Preciado-Llanes L, Napolitani G, Ashley N, Cribbs A, Koth J, Lagerholm BC, Ambrose T, Gordon MA, Sims D, Simmons A. Invasive Salmonella exploits divergent immune evasion strategies in infected and bystander dendritic cell subsets. Nat Commun 2018; 9:4883. [PMID: 30451854 PMCID: PMC6242960 DOI: 10.1038/s41467-018-07329-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/25/2018] [Indexed: 01/06/2023] Open
Abstract
Non-typhoidal Salmonella (NTS) are highly prevalent food-borne pathogens. Recently, a highly invasive, multi-drug resistant S. Typhimurium, ST313, emerged as a major cause of bacteraemia in children and immunosuppressed adults, however the pathogenic mechanisms remain unclear. Here, we utilize invasive and non-invasive Salmonella strains combined with single-cell RNA-sequencing to study the transcriptome of individual infected and bystander monocyte-derived dendritic cells (MoDCs) implicated in disseminating invasive ST313. Compared with non-invasive Salmonella, ST313 directs a highly heterogeneous innate immune response. Bystander MoDCs exhibit a hyper-activated profile potentially diverting adaptive immunity away from infected cells. MoDCs harbouring invasive Salmonella display higher expression of IL10 and MARCH1 concomitant with lower expression of CD83 to evade adaptive immune detection. Finally, we demonstrate how these mechanisms conjointly restrain MoDC-mediated activation of Salmonella-specific CD4+ T cell clones. Here, we show how invasive ST313 exploits discrete evasion strategies within infected and bystander MoDCs to mediate its dissemination in vivo.
Collapse
Affiliation(s)
- Anna Aulicino
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Kevin C Rue-Albrecht
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford, OX3 7FY, UK
| | - Lorena Preciado-Llanes
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Giorgio Napolitani
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Neil Ashley
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford and BRC Blood Theme, NIHR Oxford Biomedical Centre, Oxford, OX3 9DS, UK
| | - Adam Cribbs
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Jana Koth
- MRC Human Immunology Unit and Wolfson Imaging Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - B Christoffer Lagerholm
- MRC Human Immunology Unit and Wolfson Imaging Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Tim Ambrose
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK
| | - Melita A Gordon
- Institute of Infection and Global Health, University of Liverpool, 8 W Derby St, Liverpool, L7 3EA, UK
- Malawi-Liverpool Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - David Sims
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Alison Simmons
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
- Translational Gastroenterology Unit, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, UK.
| |
Collapse
|