1
|
Lee KM, Kim SY, Kim KH. Superior protective efficacy of genotype IVa-based single-cycle viral hemorrhagic septicemia virus (VHSV) vaccine compared to formalin-killed VHSV vaccine against VHSV genotype Ia in rainbow trout (Oncorhynchus mykiss). FISH & SHELLFISH IMMUNOLOGY 2025; 163:110409. [PMID: 40360043 DOI: 10.1016/j.fsi.2025.110409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/15/2025]
Abstract
Current viral vaccine strategies for aquaculture primarily target homologous viral infections through adaptive immune responses. However, the emergence of diverse genetic variants of viral hemorrhagic septicemia virus (VHSV) poses a significant challenge to aquaculture, necessitating vaccines capable of providing broad-spectrum cross-protection. This study aimed to compare the protective efficacy of a formalin-killed virus (FKC) vaccine and a single-cycle VHSV (rVHSV-ΔG) vaccine, both based on VHSV genotype IVa, against a heterologous VHSV genotype Ia challenge in rainbow trout (Oncorhynchus mykiss). Upon heterologous challenge with VHSV genotype Ia, the rVHSV-ΔG vaccine demonstrated superior protective efficacy relative to the FKC vaccine. Despite comparable levels of cross-reactive antibody titers and Type I interferon responses among the experimental groups, the rVHSV-ΔG group exhibited significantly elevated expression of pro-inflammatory cytokines (tnf-α, il-1β, and il-6) and the chemokine cxcl8 following challenge, suggesting an important role of innate immune mechanisms in mediating the enhanced protective effect. Notably, elevated levels of histone modifications (H3K4me3, H3K27ac) at the promoter regions of these cytokine genes suggest that rVHSV-ΔG may induce trained immunity. These results indicate that live VHSV vaccines may leverage innate immune memory to promote strong pro-inflammatory responses, representing a promising strategy for cross-protection against diverse VHSV variants.
Collapse
Affiliation(s)
- Kyung Min Lee
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 48513, South Korea
| | - So Yeon Kim
- Department of Biological Sciences, Kongju National University, Gongju, 32588, South Korea
| | - Ki Hong Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, 48513, South Korea.
| |
Collapse
|
2
|
Bonduelle O, Delory T, Franco-Moscardini I, Ghidi M, Bennacer S, Wokam M, Lenormand M, Petrier M, Rogeaux O, de Bernard S, Alves K, Nourikyan J, Lina B, INFLUOMICS Study group, Combadiere B, Janssen C. Boosting effect of high-dose influenza vaccination on innate immunity among elderly. JCI Insight 2025; 10:e184128. [PMID: 40036077 PMCID: PMC12016920 DOI: 10.1172/jci.insight.184128] [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: 07/10/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUNDThe high-dose quadrivalent influenza vaccine (QIV-HD) showed superior efficacy against laboratory-confirmed illness compared with the standard-dose quadrivalent influenza vaccine (QIV-SD) in randomized controlled trials with the elderly. However, specific underlying mechanism remains unclear.METHODSThis phase IV randomized controlled trial compared early innate responses induced by QIV-HD and QIV-SD in 59 individuals aged > 65 years. Systemic innate cells and gene signatures at day 0 (D0) and D1 as well as hemagglutinin inhibition antibody (HIA) titers at D0 and D21 after vaccination were assessed.RESULTSQIV-HD elicited robust humoral response with significantly higher antibody titers and seroconversion rates than QIV-SD. At D1 after vaccination, QIV-HD recipients showed significant reduction in innate cells, including conventional DCs and NK cells, compared with QIV-SD, correlating with significantly increased HIA titers at D21. Blood transcriptomic analysis revealed greater amplitude of gene expression in the QIV-HD arm, encompassing genes related to innate immune response, IFNs, and antigen processing and presentation, and correlated with humoral responses. Interestingly, comparative analysis with a literature dataset from young adults vaccinated with influenza standard-dose vaccine highlighted strong similarities in gene expression patterns and biological pathways with the elderly vaccinated with QIV-HD.CONCLUSIONQIV-HD induces higher HIA titers than QIV-SD, a youthful boost of the innate gene expression significantly associated with high HIA titers.TRIAL REGISTRATIONEudraCT no. 2021-004573-32.
Collapse
Affiliation(s)
- Olivia Bonduelle
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Tristan Delory
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| | - Isabelle Franco-Moscardini
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Marion Ghidi
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| | - Selma Bennacer
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Michele Wokam
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | | | - Melissa Petrier
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| | - Olivier Rogeaux
- Centre Hospitalier Centre Hospitalier Métropole Savoie, Chambéry, France
| | | | | | | | - Bruno Lina
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon-1, INSERM U1111, CNRS, UMR5308, ENS Lyon, Université Jean Monnet de Saint-Etienne, Lyon, France
- Laboratoire de Virologie, Institut des Agents Infectieux, Centre National de Référence des Virus des Infections Respiratoires, Hospices Civils de Lyon, Lyon, France
| | | | - Behazine Combadiere
- Sorbonne Université, Institut National de Santé et de Recherche Médicale, Inserm U1135, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Cécile Janssen
- Centre Hospitalier Annecy Genevois, Epagny Metz-Tessy, France
| |
Collapse
|
3
|
Bahl A, Pandey S, Rakshit R, Kant S, Tripathi D. Infection-induced trained immunity: a twist in paradigm of innate host defense and generation of immunological memory. Infect Immun 2025; 93:e0047224. [PMID: 39655962 PMCID: PMC11784091 DOI: 10.1128/iai.00472-24] [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] [Indexed: 02/01/2025] Open
Abstract
In contrast to adaptive immunity, which relies on memory T and B cells for long-term pathogen-specific responses, trained immunity involves the enhancement of innate immune responses through cellular reprogramming. Experimental evidence from animal models and human studies supports the concept of trained immunity and its potential therapeutic applications in the development of personalized medicine. However, there remains a huge gap in understanding the mechanisms, identifying specific microbial triggers responsible for the induction of trained immunity. This underscores the importance of investigating the potential role of trained immunity in redefining host defense and highlights future research directions. This minireview will provide a comprehensive summary of the new paradigm of trained immunity or innate memory pathways. It will shed light on infection-induced pathways through non-specific stimulation within macrophages and natural killer cells, which will be further elaborated in multiple disease perspectives caused by infectious agents such as bacteria, fungi, and viruses. The article further elaborates on the biochemical and cellular basis of trained immunity and its impact on disease status during recurrent exposures. The review concludes with a perspective segment discussing potential therapeutic benefits, limitations, and future challenges in this area of study. The review also sheds light upon potential risks involved in the induction of trained immunity.
Collapse
Affiliation(s)
- Aayush Bahl
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Saurabh Pandey
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, Delhi, India
| | - Roopshali Rakshit
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Sashi Kant
- Bacterial Pathogenesis, Boehringer Ingelheim Animal Health USA Inc, Ames, Iowa, USA
| | - Deeksha Tripathi
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
4
|
Das S, Lavine KJ. Role of Trained Immunity in Heath and Disease. Curr Cardiol Rep 2025; 27:18. [PMID: 39804563 DOI: 10.1007/s11886-024-02167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 05/02/2025]
Abstract
PURPOSE OF REVIEW This review aims to explore the role of immune memory and trained immunity, focusing on how innate immune cells like monocytes, macrophages, and natural killer cells undergo long-term epigenetic and metabolic rewiring. Specifically, it examines the mechanisms by which trained immunity, often triggered by infection or vaccination, could impact cardiac processes and contribute to both protective and pathological responses within the cardiovascular system. RECENT FINDINGS Recent research demonstrates that vaccination and infection not only activate immune responses in circulating monocytes and tissue macrophages but also affect immune progenitor cells within the bone marrow environment, conferring lasting protection against heterologous infections. These protective effects are attributed to epigenetic and metabolic reprogramming, which enable a heightened immune response upon subsequent encounters with pathogens. However, while trained immunity is beneficial in combating infections, it has been linked to exacerbated inflammation, which may increase susceptibility to cardiovascular diseases, including atherosclerosis and heart failure. Our review highlights the dual nature of trained immunity: while it offers protective advantages against infections, it also poses potential risks for cardiovascular health by promoting chronic inflammation. Understanding the molecular mechanisms underlying immune memory's impact on cardiac processes could lead to new therapeutic strategies to mitigate cardiovascular diseases, such as atherosclerosis, heart failure, and diabetes. These insights build the grounds for future research to balance the benefits of trained immunity with its potential risks in cardiovascular disease management.
Collapse
Affiliation(s)
- Shibali Das
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, 660 S Euclid Ave, Campus Box 8086, St. Louis, MO, 63110, USA.
| |
Collapse
|
5
|
Nyirenda J, Hardy OM, Silva Filho JD, Herder V, Attipa C, Ndovi C, Siwombo M, Namalima TR, Suwedi L, Ilia G, Nyasulu W, Ngulube T, Nyirenda D, Mvaya L, Phiri J, Chasweka D, Eneya C, Makwinja C, Phiri C, Ziwoya F, Tembo A, Makwangwala K, Khoswe S, Banda P, Morton B, Hilton O, Lawrence S, Dos Reis MF, Melo GC, de Lacerda MVG, Trindade Maranhão Costa F, Monteiro WM, Ferreira LCDL, Johnson C, McGuinness D, Jambo K, Haley M, Kumwenda B, Palmarini M, Denno DM, Voskuijl W, Kamiza SB, Barnes KG, Couper K, Marti M, Otto TD, Moxon CA. Spatially resolved single-cell atlas unveils a distinct cellular signature of fatal lung COVID-19 in a Malawian population. Nat Med 2024; 30:3765-3777. [PMID: 39567718 PMCID: PMC11645280 DOI: 10.1038/s41591-024-03354-3] [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: 11/10/2023] [Accepted: 10/14/2024] [Indexed: 11/22/2024]
Abstract
Postmortem single-cell studies have transformed understanding of lower respiratory tract diseases (LRTDs), including coronavirus disease 2019 (COVID-19), but there are minimal data from African settings where HIV, malaria and other environmental exposures may affect disease pathobiology and treatment targets. In this study, we used histology and high-dimensional imaging to characterize fatal lung disease in Malawian adults with (n = 9) and without (n = 7) COVID-19, and we generated single-cell transcriptomics data from lung, blood and nasal cells. Data integration with other cohorts showed a conserved COVID-19 histopathological signature, driven by contrasting immune and inflammatory mechanisms: in US, European and Asian cohorts, by type I/III interferon (IFN) responses, particularly in blood-derived monocytes, and in the Malawian cohort, by response to IFN-γ in lung-resident macrophages. HIV status had minimal impact on histology or immunopathology. Our study provides a data resource and highlights the importance of studying the cellular mechanisms of disease in underrepresented populations, indicating shared and distinct targets for treatment.
Collapse
Affiliation(s)
- James Nyirenda
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Olympia M Hardy
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - João Da Silva Filho
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Universität Zürich, Institut für Parasitologie, Zurich, Switzerland
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Charalampos Attipa
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Charles Ndovi
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Memory Siwombo
- Queen Elizabeth Central Hospital, Blantyre, Malawi
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | | | - Leticia Suwedi
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- International Public Health, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Georgios Ilia
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Watipenge Nyasulu
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Thokozile Ngulube
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Deborah Nyirenda
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- International Public Health, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Leonard Mvaya
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Joseph Phiri
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Dennis Chasweka
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | - Chisomo Eneya
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | | | - Chisomo Phiri
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | - Frank Ziwoya
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | - Abel Tembo
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | | | - Stanley Khoswe
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | - Peter Banda
- Queen Elizabeth Central Hospital, Blantyre, Malawi
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
| | - Ben Morton
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Orla Hilton
- Department of Infectious Diseases, Imperial College London, London, UK
| | - Sarah Lawrence
- Department of Global Health and Pediatrics, University of Washington, Seattle, WA, USA
| | - Monique Freire Dos Reis
- Department of Education and Research, Oncology Control Centre of Amazonas State (FCECON), Manaus, Brazil
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
| | | | - Marcus Vinicius Guimaraes de Lacerda
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
- Institute Leônidas & Maria Deane, Fiocruz, Manaus, Brazil
- The University of Texas Medical Branch, Galveston, TX, USA
| | | | - Wuelton Marcelo Monteiro
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Luiz Carlos de Lima Ferreira
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Carla Johnson
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dagmara McGuinness
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Kondwani Jambo
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Michael Haley
- Division of Immunology, Immunity to Infection & Respiratory Medicine, Faculty of Biology, University of Manchester, Manchester, UK
| | | | | | - Donna M Denno
- Department of Global Health and Pediatrics, University of Washington, Seattle, WA, USA
| | - Wieger Voskuijl
- Queen Elizabeth Central Hospital, Blantyre, Malawi
- Kamuzu University of Science of Health Sciences, Blantyre, Malawi
- Department of Global Health, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Kayla G Barnes
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Kevin Couper
- Division of Immunology, Immunity to Infection & Respiratory Medicine, Faculty of Biology, University of Manchester, Manchester, UK
| | - Matthias Marti
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
- Universität Zürich, Institut für Parasitologie, Zurich, Switzerland.
| | - Thomas D Otto
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Christopher A Moxon
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi.
- Malawi-Liverpool-Wellcome Programme, Kamuzu University of Health Sciences, Blantyre, Malawi.
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
6
|
Gonçalves SM, Pereira I, Feys S, Cunha C, Chamilos G, Hoenigl M, Wauters J, van de Veerdonk FL, Carvalho A. Integrating genetic and immune factors to uncover pathogenetic mechanisms of viral-associated pulmonary aspergillosis. mBio 2024; 15:e0198223. [PMID: 38651925 PMCID: PMC11237550 DOI: 10.1128/mbio.01982-23] [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] [Indexed: 04/25/2024] Open
Abstract
Invasive pulmonary aspergillosis is a severe fungal infection primarily affecting immunocompromised patients. Individuals with severe viral infections have recently been identified as vulnerable to developing invasive fungal infections. Both influenza-associated pulmonary aspergillosis (IAPA) and COVID-19-associated pulmonary aspergillosis (CAPA) are linked to high mortality rates, emphasizing the urgent need for an improved understanding of disease pathogenesis to unveil new molecular targets with diagnostic and therapeutic potential. The recent establishment of animal models replicating the co-infection context has offered crucial insights into the mechanisms that underlie susceptibility to disease. However, the development and progression of human viral-fungal co-infections exhibit a significant degree of interindividual variability, even among patients with similar clinical conditions. This observation implies a significant role for host genetics, but information regarding the genetic basis for viral-fungal co-infections is currently limited. In this review, we discuss how genetic factors known to affect either antiviral or antifungal immunity could potentially reveal pathogenetic mechanisms that predispose to IAPA or CAPA and influence the overall disease course. These insights are anticipated to foster further research in both pre-clinical models and human patients, aiming to elucidate the complex pathophysiology of viral-associated pulmonary aspergillosis and contributing to the identification of new diagnostic and therapeutic targets to improve the management of these co-infections.
Collapse
Affiliation(s)
- Samuel M Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Inês Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Simon Feys
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Georgios Chamilos
- Laboratory of Clinical Microbiology and Microbial Pathogenesis, School of Medicine, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Crete, Greece
| | - Martin Hoenigl
- Division of Infectious Diseases, ECMM Excellence Center for Medical Mycology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
| | - Joost Wauters
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
- Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Guimarães/Braga, Portugal
| |
Collapse
|
7
|
Daman AW, Cheong JG, Berneking L, Josefowicz SZ. The potency of hematopoietic stem cell reprogramming for changing immune tone. Immunol Rev 2024; 323:197-208. [PMID: 38632868 DOI: 10.1111/imr.13335] [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] [Indexed: 04/19/2024]
Abstract
Innate immune memory endows innate immune cells with antigen independent heightened responsiveness to subsequent challenges. The durability of this response can be mediated by inflammation induced epigenetic and metabolic reprogramming in hematopoietic stem and progenitor cells (HSPCs) that are maintained through differentiation to mature immune progeny. Understanding the mechanisms and extent of trained immunity induction by pathogens and vaccines, such as BCG, in HSPC remains a critical area of exploration with important implications for health and disease. Here we review these concepts and present new analysis to highlight how inflammatory reprogramming of HSPC can potently alter immune tone, including to enhance specific anti-tumor responses. New findings in the field pave the way for novel HSPC targeting therapeutic strategies in cancer and other contexts of immune modulation. Future studies are expected to unravel diverse and extensive effects of infections, vaccines, microbiota, and sterile inflammation on hematopoietic progenitor cells and begin to illuminate the broad spectrum of immunologic tuning that can be established through altering HSPC phenotypes. The purpose of this review is to draw attention to emerging and speculative topics in this field where we posit that focused study of HSPC in the framework of trained immunity holds significant promise.
Collapse
Affiliation(s)
- Andrew W Daman
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jin Gyu Cheong
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Laura Berneking
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Steven Z Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
8
|
Koekenbier EL, Fohse K, van de Maat JS, Oosterheert JJ, van Nieuwkoop C, Hoogerwerf JJ, Grobusch MP, van den Bosch MA, van de Wijgert JH, Netea MG, Rosendaal FR, Bonten MJ, Werkhoven C(HV, BCG-PRIME study group †Aardenburg-van HuisstedeAstrid9AmmerlaanHeidi S.M.10BoersmaWillem G.9BontenMarc J.M.11van den BoschMaurice A.A.J.12BrinkmanKees12Bruijning-VerhagenPatricia C.J.11van CrevelReinout13DelsingCorine14ten DoesschatenThijs11DofferhoffAnton S.M.15DuijkersRuud9FohseKonstantin13GrobuschMartin P.16GroenwoldRolf H.H.17de HaasCorine11HassingRobert-Jan18de HoogMarieke L.A.11HoogerwerfJacobien J.13HuijtsSusanne M.19van Hylckama-VliegAstrid17JongEefje20de JongHanna K.21KnapMartijn21KoekenbierEva L.11KoendersMichael22KouijzerIlse J.E.13KramerHenk23van de LaarRoel24LalmohamedArief11LensenKarel-Jan D.F.25LijferingWillem M.17van de MaatJosephine S.13MagdelijnsFabienne26MeekBob27MiddelburgRutger A.17MoeniralamHazra S.27MooijaartSimon P.17van MunsterBarbara C.25NeteaMihai G.13van NieuwkoopCees28ten OeverJaap13OosterheertJan Jelrik11GoossensMarc Padros11PetersVincent10PostmaDouwe F.25PouwNiels13ReesinkHerre J.12de RegtMarieke J.A.12van der ReijdenAnneli C.J.29RosendaalFrits R.17SchaakxsR.11SliekerKitty30SlingerlandRobbert J.31van SluisNicolette L.J.11StehouwerCoen D.A.26van de VeerdonkFrank13VerbonAnnelies19van WerkhovenC.H. (Henri)11van de WijgertJanneke H.H.11Noordwest Ziekenhuis, Alkmaar, the NetherlandsCatharina Ziekenhuis, Eindhoven, the NetherlandsUniversity Medical Center Utrecht, Utrecht, the NetherlandsOLVG, Amsterdam, the NetherlandsRadboud University Medical Center, Nijmegen, the NetherlandsMedisch Spectrum Twente, Enschede, the NetherlandsCanisius-Wilhelmina Hospital, NijmegenAmsterdam UMC, Amsterdam, the NetherlandsLeiden University Medical Center, Leiden, the NetherlandsRijnstate Hospital, Arnhem, the NetherlandsErasmus Medical Center, Rotterdam, the NetherlandsMeander Medisch Centrum, Amersfoort, the NetherlandsUniversity Medical Center Amsterdam, Amsterdam, the NetherlandsSanteon Bureau of Research & Innovation, Utrecht, the NetherlandsMartini Hospital, Groningen, the NetherlandsIkazia Hospital, Rotterdam, the NetherlandsUniversity Medical Center Groningen, Groningen, the NetherlandsUniversity Medical Center Maastricht, Maastricht, the NetherlandsSt. Antonius Hospital, Nieuwegein, the NetherlandsHaga Teaching Hospital, The Hague, the NetherlandsAnticoagulation Clinic Leiden, Leiden, the NetherlandsBernhoven Hospital, Uden, the NetherlandsMaasstad Hospital, Rotterdam, the Netherlands, Werkhoven C(HV, BCG-PRIME study group. Bacillus Calmette-Guérin vaccine for prevention of COVID-19 and other respiratory tract infections in older adults with comorbidities: a randomized controlled trial. Clin Microbiol Infect 2023:S1198-743X(23)00044-7. [PMID: 36736662 PMCID: PMC9892323 DOI: 10.1016/j.cmi.2023.01.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To test whether Bacillus Calmette-Guérin (BCG) vaccination would reduce the incidence of COVID-19 and other respiratory tract infections (RTIs) in older adults with one or more comorbidities. METHODS Community-dwelling adults aged 60 years or older with one or more underlying comorbidities and no contraindications to BCG vaccination were randomized 1:1 to BCG or placebo vaccination and followed for 6 months. The primary endpoint was a self-reported, test-confirmed COVID-19 incidence. Secondary endpoints included COVID-19 hospital admissions and clinically relevant RTIs (i.e. RTIs including but not limited to COVID-19 requiring medical intervention). COVID-19 and clinically relevant RTI episodes were adjudicated. Incidences were compared using Fine-Gray regression, accounting for competing events. RESULTS A total of 6112 participants with a median age of 69 years (interquartile range, 65-74) and median of 2 (interquartile range, 1-3) comorbidities were randomized to BCG (n = 3058) or placebo (n = 3054) vaccination. COVID-19 infections were reported by 129 BCG recipients compared to 115 placebo recipients [hazard ratio (HR), 1.12; 95% CI, 0.87-1.44]. COVID-19-related hospitalization occurred in 18 BCG and 21 placebo recipients (HR, 0.86; 95% CI, 0.46-1.61). During the study period, 13 BCG recipients died compared with 18 placebo recipients (HR, 0.71; 95% CI, 0.35-1.43), of which 11 deaths (35%) were COVID-19-related: six in the placebo group and five in the BCG group. Clinically relevant RTI was reported by 66 BCG and 72 placebo recipients (HR, 0.92; 95% CI, 0.66-1.28). DISCUSSION BCG vaccination does not protect older adults with comorbidities against COVID-19, COVID-19 hospitalization, or clinically relevant RTIs.
Collapse
Affiliation(s)
- Eva L. Koekenbier
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands,Corresponding author. Eva L. Koekenbier, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Postbus 85500, 3584 GC Utrecht, the Netherlands
| | - Konstantin Fohse
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Josephine S. van de Maat
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Jan Jelrik Oosterheert
- Department of Infectious Diseases, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Cees van Nieuwkoop
- Department of Internal Medicine, Haga Teaching Hospital, The Hague, the Netherlands
| | - Jacobien J. Hoogerwerf
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Martin P. Grobusch
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Infection & Immunity, Amsterdam Public Health, Amsterdam, the Netherlands
| | | | - Janneke H.H. van de Wijgert
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Centre, Nijmegen, the Netherlands,Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Germany
| | - Frits R. Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marc J.M. Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - C.H. (Henri) van Werkhoven
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - BCG-PRIME study group†Aardenburg-van HuisstedeAstrid9AmmerlaanHeidi S.M.10BoersmaWillem G.9BontenMarc J.M.11van den BoschMaurice A.A.J.12BrinkmanKees12Bruijning-VerhagenPatricia C.J.11van CrevelReinout13DelsingCorine14ten DoesschatenThijs11DofferhoffAnton S.M.15DuijkersRuud9FohseKonstantin13GrobuschMartin P.16GroenwoldRolf H.H.17de HaasCorine11HassingRobert-Jan18de HoogMarieke L.A.11HoogerwerfJacobien J.13HuijtsSusanne M.19van Hylckama-VliegAstrid17JongEefje20de JongHanna K.21KnapMartijn21KoekenbierEva L.11KoendersMichael22KouijzerIlse J.E.13KramerHenk23van de LaarRoel24LalmohamedArief11LensenKarel-Jan D.F.25LijferingWillem M.17van de MaatJosephine S.13MagdelijnsFabienne26MeekBob27MiddelburgRutger A.17MoeniralamHazra S.27MooijaartSimon P.17van MunsterBarbara C.25NeteaMihai G.13van NieuwkoopCees28ten OeverJaap13OosterheertJan Jelrik11GoossensMarc Padros11PetersVincent10PostmaDouwe F.25PouwNiels13ReesinkHerre J.12de RegtMarieke J.A.12van der ReijdenAnneli C.J.29RosendaalFrits R.17SchaakxsR.11SliekerKitty30SlingerlandRobbert J.31van SluisNicolette L.J.11StehouwerCoen D.A.26van de VeerdonkFrank13VerbonAnnelies19van WerkhovenC.H. (Henri)11van de WijgertJanneke H.H.11Noordwest Ziekenhuis, Alkmaar, the NetherlandsCatharina Ziekenhuis, Eindhoven, the NetherlandsUniversity Medical Center Utrecht, Utrecht, the NetherlandsOLVG, Amsterdam, the NetherlandsRadboud University Medical Center, Nijmegen, the NetherlandsMedisch Spectrum Twente, Enschede, the NetherlandsCanisius-Wilhelmina Hospital, NijmegenAmsterdam UMC, Amsterdam, the NetherlandsLeiden University Medical Center, Leiden, the NetherlandsRijnstate Hospital, Arnhem, the NetherlandsErasmus Medical Center, Rotterdam, the NetherlandsMeander Medisch Centrum, Amersfoort, the NetherlandsUniversity Medical Center Amsterdam, Amsterdam, the NetherlandsSanteon Bureau of Research & Innovation, Utrecht, the NetherlandsMartini Hospital, Groningen, the NetherlandsIkazia Hospital, Rotterdam, the NetherlandsUniversity Medical Center Groningen, Groningen, the NetherlandsUniversity Medical Center Maastricht, Maastricht, the NetherlandsSt. Antonius Hospital, Nieuwegein, the NetherlandsHaga Teaching Hospital, The Hague, the NetherlandsAnticoagulation Clinic Leiden, Leiden, the NetherlandsBernhoven Hospital, Uden, the NetherlandsMaasstad Hospital, Rotterdam, the Netherlands
| | - C.H. (Henri) van Werkhoven
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | |
Collapse
|
9
|
Föhse K, Taks EJM, Moorlag SJCFM, Bonten MJM, van Crevel R, Ten Oever J, van Werkhoven CH, Netea MG, van de Maat JS, Hoogerwerf JJ. The impact of circadian rhythm on Bacillus Calmette-Guérin vaccination effects on SARS-CoV-2 infections. Front Immunol 2023; 14:980711. [PMID: 36875134 PMCID: PMC9978461 DOI: 10.3389/fimmu.2023.980711] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Background and objective A recent study has suggested that circadian rhythm has an important impact on the immunological effects induced by Bacillus Calmette-Guérin (BCG) vaccination. The objective of this study was to evaluate whether the timing of BCG vaccination (morning or afternoon) affects its impact on severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infections and clinically relevant respiratory tract infections (RTIs). Methods This is a post-hoc analysis of the BCG-CORONA-ELDERLY (NCT04417335) multicenter, placebo-controlled trial, in which participants aged 60 years and older were randomly assigned to vaccination with BCG or placebo, and followed for 12 months. The primary endpoint was the cumulative incidence of SARS-CoV-2 infection. To assess the impact of circadian rhythm on the BCG effects, participants were divided into four groups: vaccinated with either BCG or placebo in the morning (between 9:00h and 11:30h) or in the afternoon (between 14:30h and 18:00h). Results The subdistribution hazard ratio of SARS-CoV-2 infection in the first six months after vaccination was 2.394 (95% confidence interval [CI], 0.856-6.696) for the morning BCG group and 0.284 (95% CI, 0.055-1.480) for the afternoon BCG group. When comparing those two groups, the interaction hazard ratio was 8.966 (95% CI, 1.366-58.836). In the period from six months until 12 months after vaccination cumulative incidences of SARS-CoV-2 infection were comparable, as well as cumulative incidences of clinically relevant RTI in both periods. Conclusion Vaccination with BCG in the afternoon offered better protection against SARS-CoV-2 infections than BCG vaccination in the morning in the first six months after vaccination.
Collapse
Affiliation(s)
- Konstantin Föhse
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Esther J M Taks
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Simone J C F M Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marc J M Bonten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jaap Ten Oever
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cornelis H van Werkhoven
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Josephine S van de Maat
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jacobien J Hoogerwerf
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
10
|
Do bacterial vaccines/adjuvants prevent wheezing episodes in children? Curr Opin Allergy Clin Immunol 2022; 22:380-386. [PMID: 36305468 DOI: 10.1097/aci.0000000000000854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
PURPOSE OF REVIEW To discuss recently discovered mechanisms of action of some bacterial vaccines that may account for their clinical benefit in the prevention of recurrent wheezing and asthma exacerbations in infants and early childhood. RECENT FINDINGS Trained immunity has been shown to confer innate immune cells with a quite long-term nonspecific protection against a broad spectrum of pathogens. Inducers of trained immunity include some bacterial vaccines. Trained immunity-based vaccines (TIbV) of bacterial origin have the capability to induce nonspecific responses to a variety of pathogens, including respiratory viruses, in addition to their nominal bacterial antigens. Clinical data, from epidemiological surveys to well designed randomized clinical trials, indicate that TIbV formulated with bacteria prevent respiratory tract infections of viral cause, such as those associated with recurrent wheezing or asthma exacerbation, in children. Administration of these vaccines by the mucosal route may be important for their outcome in respiratory infections. SUMMARY Mucosal bacterial immunotherapy, including certain TIbV, confer protection against a broad spectrum of pathogens, such as viruses, through a mechanism mediated by trained immunity. Clinical studies on the use of these preparations against recurrent wheezing reflect these mechanistic effects. These findings open a new avenue for the development of new strategies for this condition.
Collapse
|