1
|
Ghimire R, Shrestha R, Amaradhi R, Liu L, More S, Ganesh T, Ford AK, Channappanavar R. Toll-like receptor 7 (TLR7)-mediated antiviral response protects mice from lethal SARS-CoV-2 infection. J Virol 2025; 99:e0166824. [PMID: 40162785 DOI: 10.1128/jvi.01668-24] [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] [Received: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced impaired antiviral immunity and excessive inflammatory responses cause lethal pneumonia. However, the in vivo roles of key pattern recognition receptors that elicit protective antiviral and fatal inflammatory responses, specifically in the lungs, are not well described. Coronaviruses possess single-stranded RNA genome that activates TLR7/8 to induce an antiviral interferon (IFN) and robust inflammatory cytokine response. Here, using wild-type and TLR7-deficient (TLR7-/-) mice infected with mouse-adapted SARS-CoV-2 (MA-CoV-2), we examined the role of TLR7 in the lung antiviral and inflammatory response and severe pneumonia. We showed that TLR7 deficiency significantly increased lung virus loads and morbidity/mortality, which correlated with reduced levels of type I IFNs (Ifna/b), type III IFNs (Ifnl), and IFN-stimulated genes (ISGs) in the lungs. A detailed evaluation of MA-CoV-2-infected lungs revealed increased neutrophil accumulation and lung pathology in TLR7-/- mice. We further showed that blocking type I IFN receptor (IFNAR) signaling enhanced SARS-CoV-2 replication in the lungs and caused severe lung pathology, leading to 100% mortality compared to infected control mice. Moreover, immunohistochemical assessment of the lungs revealed increased numbers of SARS-CoV-2 antigen-positive macrophages, pneumocytes, and bronchial epithelial cells in TLR7-/- and IFNAR-deficient mice compared to control mice. In summary, we conclusively demonstrated that despite TLR7-induced robust lung inflammation, TLR7-induced IFN/ISG responses suppress lung virus replication and pathology and provide protection against SARS-CoV-2-induced fatal pneumonia. Additionally, given the similar disease outcomes in control, TLR7-/-, and IFNAR-deficient MA-CoV-2-infected mice and coronavirus disease 2019 (COVID-19) patients, we propose that MA-CoV-2-infected mice constitute an excellent model for studying COVID-19.IMPORTANCESevere coronavirus disease 2019 (COVID-19) is caused by a delicate balance between a strong antiviral and an exuberant inflammatory response. A robust antiviral immunity and regulated inflammation are protective, while a weak antiviral response and excessive inflammation are detrimental. However, the key host immune sensors that elicit protective antiviral and inflammatory responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge are poorly defined. Here, we examined the role of viral RNA-mediated TLR7 activation in the lung antiviral and inflammatory responses in SARS-CoV-2-infected mice. We demonstrate that TLR7 deficiency led to a high rate of morbidity and mortality, which correlated with an impaired antiviral interferon (IFN)-I/III response, enhanced lung virus replication, and severe lung pathology. Furthermore, we show that blocking IFN-I signaling using anti-IFN receptor antibody promoted SARS-CoV-2 replication in the lungs and caused severe disease. These results provide conclusive evidence that TLR7 and IFN-I receptor deficiencies lead to severe disease in mice, replicating clinical features observed in COVID-19 patients.
Collapse
Affiliation(s)
- Roshan Ghimire
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rakshya Shrestha
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lin Liu
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alexandra K Ford
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rudragouda Channappanavar
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
2
|
Chaumont A, Martin A, Flamaing J, Wiseman DJ, Vandermeulen C, Jongert E, Doherty TM, Buchy P, Varga SM, Warter L. Host immune response to respiratory syncytial virus infection and its contribution to protection and susceptibility in adults: a systematic literature review. Expert Rev Clin Immunol 2025:1-16. [PMID: 40278893 DOI: 10.1080/1744666x.2025.2494658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/26/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
INTRODUCTION Respiratory syncytial virus (RSV) is an important pathogen in infants, children, older adults, and those with comorbidities. Mechanisms involving viral proteins appear to underlie the ability of RSV to evade and modulate host immunity. We aimed to understand virus- and host-dependent factors regulating the development and severity of RSV infection, as related to the prevention and treatment of RSV-associated disease in adults, through a systematic literature review (SLR). METHODS An SLR was conducted to identify immune mechanisms involved in the protective response to RSV infection in adults, and responses that may contribute to the development of severe disease. Concurrent searches (MEDLINE/Embase) using embase.com identified relevant papers published between 1990 and 19 April 2023. RESULTS Of 1813 records identified, 113 were selected for review. Inclusion criteria were based on relevant patient populations, outcomes, and study methodologies. RSV is common, recurrent, and associated with high morbidity and mortality in older adults and people with underlying chronic diseases. Immune responses differ between younger and older adults. The approval of effective vaccines may protect older individuals from symptomatic RSV infection. CONCLUSIONS We established the complexities of RSV immune response, but further research is required to fully understand anti-RSV immunology.
Collapse
Affiliation(s)
| | | | - Johan Flamaing
- Department of Geriatric Medicine, University Hospitals Leuven, Leuven, Belgium
- Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Dexter J Wiseman
- Department of Respiratory Medicine, Chelsea and Westminster Hospital, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | | | | | - Steven M Varga
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
3
|
Norris PAA, Kubes P. Innate immunity of the lungs in homeostasis and disease. Mucosal Immunol 2025:S1933-0219(25)00039-X. [PMID: 40220792 DOI: 10.1016/j.mucimm.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/30/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
Humans breathe thousands of litres of non-sterile air each day containing bacteria, viruses, and fungi, as well as pollutants, allergens, and other particles. The continual exposure to foreign particles is juxtaposed with the vast surface area of the blood-air-barrier which becomes extremely thin to allow for efficient gas exchange. To prevent infection and injury, the healthy lung relies on a robust innate immune system to protect itself. Critically, this innate immune system must clear insults while maintaining immune tolerance and minimizing inflammation to avoid disrupting the lung's vital gas exchange function. In this review, we discuss how the innate immune system protects the lung from its environment.
Collapse
Affiliation(s)
- Peter A A Norris
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Paul Kubes
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
4
|
Sagawe JS, Loake VIP, Openshaw PJM, Kemp P, Culley FJ. Aging enhances pro-atrogenic gene expression and skeletal muscle loss following respiratory syncytial virus infection. GeroScience 2025; 47:1485-1500. [PMID: 39354240 PMCID: PMC11978595 DOI: 10.1007/s11357-024-01370-2] [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: 07/12/2024] [Accepted: 09/25/2024] [Indexed: 10/03/2024] Open
Abstract
Aging and many age-related health conditions are associated with skeletal muscle loss. Furthermore, older adults are more susceptible to severe respiratory infections, which can in turn lead to muscle wasting. The mechanisms by which respiratory viral infection can impact skeletal muscle in older adults are not well understood. We determined the effects of acute infection with respiratory syncytial virus (RSV) on the lung and skeletal muscle of aged mice. RSV infection caused more severe disease in aged mice with enhanced weight loss, reduced feeding, higher viral load, and greater airway inflammation. Aged but not young mice showed decreased leg muscle weight at the peak of illness and decreased size of leg muscle fibers. Aged mice increased muscle-specific expression of atrophy-promoting enzymes (Atrogin-1 and MuRF-1) and failed to increase the rate of muscle protein synthesis during RSV infection. In aged mice, the changes in Atrogin-1 and MuRF-1 gene expression in skeletal muscle correlated with IL-6 levels in the lungs. These findings indicate that RSV infection of aged mice provides a model for studying the diverse adverse systemic consequences of respiratory viral infections on health and wellbeing in older adults.
Collapse
Affiliation(s)
- J Sophie Sagawe
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Verity I P Loake
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Peter J M Openshaw
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Paul Kemp
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Fiona J Culley
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, St Mary's Campus, Norfolk Place, London, W2 1PG, UK.
| |
Collapse
|
5
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
6
|
Lim PN, Cervantes MM, Pham LK, Doherty SR, Tufts A, Dubey D, Mai D, Aderem A, Diercks AH, Rothchild AC. Absence of c-Maf and IL-10 enables type I IFN enhancement of innate responses to LPS in alveolar macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae029. [PMID: 40073087 PMCID: PMC11952875 DOI: 10.1093/jimmun/vkae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/19/2024] [Indexed: 03/14/2025]
Abstract
Alveolar macrophages (AMs) are lung-resident myeloid cells and airway sentinels for inhaled pathogens and environmental particles. While AMs can be highly inflammatory in response to respiratory viruses, they do not mount proinflammatory responses to all airborne pathogens. For example, we previously showed that AMs fail to mount a robust proinflammatory response to Mycobacterium tuberculosis. Here, we address this discrepancy by investigating the capacity of murine AMs for direct innate immune sensing, using LPS as a model. Use of LPS-coated fluorescent beads enabled us to distinguish between directly exposed and bystander cells to measure transcriptional responses, by RNA-sequencing after cell sorting, and cytokine responses, by flow cytometry. We find that AMs have decreased proinflammatory responses to low-dose LPS compared to other macrophage types (bone marrow-derived macrophages, peritoneal macrophages), as measured by TNF, IL-6, Ifnb, and Ifit3. The reduced response to low-dose LPS correlates with minimal TLR4 and CD14 surface expression, despite sufficient internal expression of TLR4. We also find that AMs do not produce IL-10 in response to a variety of stimuli due to low expression of the transcription factor c-Maf, while exogenous c-Maf expression restores IL-10 production in AMs. Lastly, we show that lack of IL-10 enables type I IFN enhancement of AM responses to LPS. Overall, we demonstrate AMs have a cell-intrinsic hyporesponsiveness to LPS, which makes them uniquely tolerant to low-dose exposure. Regulation of AM innate responses by distinct CD14, c-Maf, and IL-10 expression patterns has important implications for both respiratory infections and environmental airborne exposures.
Collapse
Affiliation(s)
- Pamelia N Lim
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| | - Maritza M Cervantes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Linh K Pham
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Animal Biotechnology & Biomedical Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Sydney R Doherty
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ankita Tufts
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Divya Dubey
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| | - Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alan H Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alissa C Rothchild
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
7
|
Li S, Pan M, Zhao H, Li Y. Role of CCL2/CCR2 axis in pulmonary fibrosis induced by respiratory viruses. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00036-2. [PMID: 39955168 DOI: 10.1016/j.jmii.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 01/23/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Respiratory virus infection is an important cause of both community acquired pneumonia and hospital-acquired pneumonia. Various respiratory viruses, including influenza virus, avian influenza virus, respiratory syncytial virus (RSV), SARS-CoV, MERS-CoV, and SARS-CoV-2, result in severe fibrosis sequelae after the acute phase. Since the COVID-19 pandemic, respiratory virus infection, as an important cause of pulmonary fibrosis, has attracted increasing attention around the world. Respiratory virus infection usually triggers robust inflammation responses, leading to large amounts of proinflammatory mediator production, such as chemokine (C-C motif) ligand 2 (CCL2), a critical chemokine involved in the recruitment of various inflammatory cells. Moreover, CCL2 plays a pivotal role in the pathogenesis of fibrosis progression, through regulating recruitment of bone marrow-derived monocytes and increasing the expression of extracellular matrix proteins. This review provided a concise overview of the common fibrosis sequelae after virus infection. Then we discussed the elevated levels of CCL2 in various respiratory virus infection, underscoring its potent profibrotic role. Targeting the CCL2/CCR2 axis holds promise for alleviating fibrosis sequelae post-acute virus infection and warrants further investigation.
Collapse
Affiliation(s)
- Shuangyan Li
- Beijing Hospital, National Centre of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730, Beijing, China.
| | - Mingming Pan
- Department of Respiratory and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| | - Hui Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, 100071, Beijing, China.
| | - Yanming Li
- Department of Respiratory and Critical Care Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| |
Collapse
|
8
|
Miller JL, Niewiesk S. Review of impaired immune parameters in RSV infections in the elderly. Virology 2025; 603:110395. [PMID: 39827596 DOI: 10.1016/j.virol.2025.110395] [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: 10/17/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Respiratory syncytial virus (RSV) infections in elderly individuals are associated with increased rates of severe clinical disease and mortality compared to younger adults. Age-associated declines in numerous innate and adaptive immune parameters during RSV infection contribute to infection susceptibility, impaired viral clearance, and distorted cytokine profiles in the elderly. Impaired immune responses in this age group also adversely affect longevity of RSV immunity following vaccination in experimental settings. This review summarizes the effects of aging on cellular immune responses to RSV in humans and animal models, molecular mechanisms for these impaired responses where they have been elucidated, and the clinical consequences of impaired immunity in the elderly.
Collapse
Affiliation(s)
- Jonathan L Miller
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
9
|
Zheng Y, Corrêa-Silva S, Rodrigues RM, Corrêa de Souza E, Macaferri da Fonseca FA, Gilio AE, Carneiro-Sampaio M, Palmeira P. Infant respiratory infections modulate lymphocyte homing to breast milk. Front Immunol 2025; 15:1481416. [PMID: 39867906 PMCID: PMC11757141 DOI: 10.3389/fimmu.2024.1481416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/16/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Chemokines and their receptors are essential for leukocyte migration to several tissues, including human milk. Here, we evaluated the homing of T and B lymphocyte subsets to breast milk in response to ongoing respiratory infections in the nursing infant. Methods Blood and mature milk were collected from healthy mothers of nurslings with respiratory infections (Group I) and from healthy mothers of healthy nurslings (Group C). Total lymphocyte, T and B cells, their subset numbers, and the expression of the homing receptors CCR5, CCR6, CCR10, and CXCR3 in these cells were evaluated in milk. Maternal serum and milk chemokine, cytokine, and IgA and IgG antibody levels were also quantified. Results All milk lymphocyte numbers were greater in Group I than in Group C. All CD4 T-cell subsets expressing CCR5, CCR6, and CXCR3 were higher in Group I. Within the CD8 T-cell subsets, only CCR6 and CXCR3 were higher in Group I, while CCR5 expression was higher in Group I exclusively for activated CD8 T cells. Group I showed greater numbers of all CCR6+ B-cell subsets and CXCR3+ naive B cells and plasma cells than did Group C. Infection of the nurslings promoted increased CCL20, CXCL10, IL-6, IL-8, total IgA, and IgG levels in the milk. Conclusion Respiratory infections in nursing infants stimulate an increase in cytokines and chemokines in breast milk, facilitating the recruitment and activation of lymphocytes. This process may promote immunological tolerance and help in the maturation of the infant's immune system, providing an additional strategy for passive maternal-infant protection.
Collapse
Affiliation(s)
- Yingying Zheng
- Department of Pediatrics, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Simone Corrêa-Silva
- Laboratorio de Pediatria Clinica (LIM36), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Regina Maria Rodrigues
- Department of Pediatrics, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Eloisa Corrêa de Souza
- Department of Pediatrics, University Hospital, Medical School, University of São Paulo, São Paulo, Brazil
| | | | - Alfredo Elias Gilio
- Department of Pediatrics, University Hospital, Medical School, University of São Paulo, São Paulo, Brazil
| | - Magda Carneiro-Sampaio
- Department of Pediatrics, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Patricia Palmeira
- Laboratorio de Pediatria Clinica (LIM36), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
10
|
Pöpperl P, Stoff M, Beineke A. Alveolar Macrophages in Viral Respiratory Infections: Sentinels and Saboteurs of Lung Defense. Int J Mol Sci 2025; 26:407. [PMID: 39796262 PMCID: PMC11721917 DOI: 10.3390/ijms26010407] [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: 12/09/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Respiratory viral infections continue to cause pandemic and epidemic outbreaks in humans and animals. Under steady-state conditions, alveolar macrophages (AlvMϕ) fulfill a multitude of tasks in order to maintain tissue homeostasis. Due to their anatomic localization within the deep lung, AlvMϕ are prone to detect and react to inhaled viruses and thus play a role in the early pathogenesis of several respiratory viral infections. Here, detection of viral pathogens causes diverse antiviral and proinflammatory reactions. This fact not only makes them promising research targets, but also suggests them as potential targets for therapeutic and prophylactic approaches. This review aims to give a comprehensive overview of the current knowledge about the role of AlvMϕ in respiratory viral infections of humans and animals.
Collapse
Affiliation(s)
- Pauline Pöpperl
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| | - Melanie Stoff
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Center for Systems Neuroscience (ZSN), 30559 Hannover, Germany
| |
Collapse
|
11
|
Duan Y, Liu Z, Zang N, Cong B, Shi Y, Xu L, Jiang M, Wang P, Zou J, Zhang H, Feng Z, Feng L, Ren L, Liu E, Li Y, Zhang Y, Xie Z. Landscape of respiratory syncytial virus. Chin Med J (Engl) 2024; 137:2953-2978. [PMID: 39501814 PMCID: PMC11706595 DOI: 10.1097/cm9.0000000000003354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Indexed: 01/11/2025] Open
Abstract
ABSTRACT Respiratory syncytial virus (RSV) is an enveloped, negative-sense, single-stranded RNA virus of the Orthopneumovirus genus of the Pneumoviridae family in the order Mononegavirales. RSV can cause acute upper and lower respiratory tract infections, sometimes with extrapulmonary complications. The disease burden of RSV infection is enormous, mainly affecting infants and older adults aged 75 years or above. Currently, treatment options for RSV are largely supportive. Prevention strategies remain a critical focus, with efforts centered on vaccine development and the use of prophylactic monoclonal antibodies. To date, three RSV vaccines have been approved for active immunization among individuals aged 60 years and above. For children who are not eligible for these vaccines, passive immunization is recommended. A newly approved prophylactic monoclonal antibody, Nirsevimab, which offers enhanced neutralizing activity and an extended half-life, provides exceptional protection for high-risk infants and young children. This review provides a comprehensive and detailed exploration of RSV's virology, immunology, pathogenesis, epidemiology, clinical manifestations, treatment options, and prevention strategies.
Collapse
Affiliation(s)
- Yuping Duan
- School of Population Medicine and Public Health, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Zimeng Liu
- National Health Commission Key Laboratory of Systems Biology of Pathogen, Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102629, China
| | - Na Zang
- Department of Respiratory Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Key Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing 400014, China
| | - Bingbing Cong
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yuqing Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Medical Virology and Viral Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Lili Xu
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences (2019RU016), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health,Beijing 100045, China
| | - Mingyue Jiang
- School of Population Medicine and Public Health, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Peixin Wang
- National Health Commission Key Laboratory of Systems Biology of Pathogen, Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102629, China
| | - Jing Zou
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Han Zhang
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ziheng Feng
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences (2019RU016), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health,Beijing 100045, China
| | - Luzhao Feng
- School of Population Medicine and Public Health, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lili Ren
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
- National Health Commission Key Laboratory of Systems Biology of Pathogen, Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102629, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Enmei Liu
- Department of Respiratory Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Key Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing 400014, China
| | - You Li
- Department of Epidemiology, National Vaccine Innovation Platform, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, UK
- Changzhou Third People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu 213000, China
| | - Yan Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, NHC Key Laboratory of Medical Virology and Viral Disease, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Zhengde Xie
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences (2019RU016), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health,Beijing 100045, China
| |
Collapse
|
12
|
Gao S, Li W, Huang Z, Deiuliis JA, Braunstein Z, Liu X, Li X, Kosari M, Chen J, Min X, Yang H, Gong Q, Liu Z, Wei Y, Zhang Z, Dong L, Zhong J. Deciphering the therapeutic potential of Myeloid-Specific JAK2 inhibition in acute respiratory distress syndrome. Mucosal Immunol 2024; 17:1273-1284. [PMID: 39173745 DOI: 10.1016/j.mucimm.2024.08.008] [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: 11/06/2023] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening condition characterized by severe inflammation and pulmonary dysfunction. Despite advancements in critical care, effective pharmacological interventions for ARDS remain elusive. While Janus kinase 2 (JAK2) inhibitors have emerged as an innovative treatment for numerous autoinflammatory diseases, their therapeutic potential in ARDS remains unexplored. In this study, we investigated the contribution of JAK2 and its underlying mechanisms in ARDS utilizing myeloid-specific JAK2 knockout murine models alongside a pharmacological JAK2 inhibitor. Notably, myeloid-specific JAK2 knockout led to a notable attenuation of ARDS induced by intratracheal administration of LPS, accompanied by reduced levels of neutrophils and inflammatory cytokines in bronchoalveolar lavage fluid (BALF) and lung tissue. Intriguingly, the ameliorative effects were abolished upon the depletion of monocyte-derived alveolar macrophages (Mo-AMs) rather than tissue-resident alveolar macrophages (TR-AMs). JAK2 deficiency markedly reversed LPS-induced activation of STAT5 in macrophages. Remarkably, pharmacological JAK2 inhibition using baricitinib failed to substantially alleviate neutrophils infiltration, implying that specific inhibition of JAK2 in Mo-AMs is imperative for ARDS amelioration. Collectively, our data suggest that JAK2 may mitigate ARDS progression through the JAK2 pathway in Mo-AMs, underscoring JAK2 in alveolar macrophages, particularly Mo-AMs, as a promising therapeutic target for ARDS treatment.
Collapse
Affiliation(s)
- Shupei Gao
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wenjuan Li
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhiwen Huang
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jeffrey A Deiuliis
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zachary Braunstein
- Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Xinxin Liu
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xinlu Li
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Mohammadreza Kosari
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jun Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, Hubei 442008, China
| | - Xinwen Min
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, Hubei 442008, China
| | - Handong Yang
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research (Hubei University of Medicine), Shiyan, Hubei 442008, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei 434023, China
| | - Zheng Liu
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ziyang Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan 430030, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA; Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, Hubei 430030, China.
| |
Collapse
|
13
|
Panahipoor Javaherdehi A, Ghanbari S, Mahdavi P, Zafarani A, Razizadeh MH. The role of alveolar macrophages in viral respiratory infections and their therapeutic implications. Biochem Biophys Rep 2024; 40:101826. [PMID: 39324036 PMCID: PMC11422589 DOI: 10.1016/j.bbrep.2024.101826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
Alveolar macrophages are pivotal components of the lung's innate immune defense against respiratory virus infections. Their multifaceted role spans from viral clearance to modulation of immune responses, making them essential players in shaping disease outcomes. In this comprehensive review collection, we look into the intricate interplay between Alveolar macrophages and various respiratory viruses, shedding light on their dynamic contributions to immune resilience. From influenza to respiratory syncytial virus, Alveolar macrophages emerge as sentinels of the airways, actively participating in viral detection and initiating rapid antiviral responses. Their ability to recognize viral pathogens triggers a cascade of events, including cytokine and chemokine production that guides the recruitment and activation of immune effectors. Furthermore, Alveolar macrophages impact the fate of adaptive immune responses by modulating the activation of T lymphocytes and the secretion of key cytokines. These reviews encompass a range of insights, including the regulation of inflammasome activation, the influence of Alveolar macrophages on cytokine dysregulation, and their role in preventing secondary bacterial pneumonia post-infection. Collectively, they highlight the significance of Alveolar macrophages in preserving pulmonary integrity and immune homeostasis during viral challenges.
Collapse
Affiliation(s)
| | | | - Pooya Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Zafarani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Razizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Girma A. Biology of human respiratory syncytial virus: Current perspectives in immune response and mechanisms against the virus. Virus Res 2024; 350:199483. [PMID: 39396572 PMCID: PMC11513633 DOI: 10.1016/j.virusres.2024.199483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Human respiratory syncytial virus (hRSV) remains a leading cause of morbidity and mortality in infants, young children, and older adults. hRSV infection's limited treatment and vaccine options significantly increase bronchiolitis' morbidity rates. The severity and outcome of viral infection hinge on the innate immune response. Developing vaccines and identifying therapeutic interventions suitable for young children, older adults, and pregnant women relies on comprehending the molecular mechanisms of viral PAMP recognition, genetic factors of the inflammatory response, and antiviral defense. This review covers fundamental elements of hRSV biology, diagnosis, pathogenesis, and the immune response, highlighting prospective options for vaccine development.
Collapse
Affiliation(s)
- Abayeneh Girma
- Department of Biology, College of Natural and Computational Sciences, Mekdela Amba University, P.O. Box 32, Tulu Awuliya, Ethiopia.
| |
Collapse
|
15
|
Owen AR, Farias A, Levins AM, Wang Z, Higham SL, Mack M, Tregoning JS, Johansson C. Exposure to bacterial PAMPs before RSV infection exacerbates innate inflammation and disease via IL-1α and TNF-α. Mucosal Immunol 2024; 17:1184-1198. [PMID: 39127259 PMCID: PMC11631774 DOI: 10.1016/j.mucimm.2024.08.002] [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: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Respiratory syncytial virus (RSV) can cause severe lower respiratory tract infections. Understanding why some individuals get more serious disease may help with diagnosis and treatment. One possible risk factor underlying severe disease is bacterial exposure before RSV infection. Bacterial exposure has been associated with increased respiratory viral-induced disease severity but the mechanism remains unknown. Respiratory bacterial infections or exposure to their pathogen associated molecular patterns (PAMPs) trigger innate immune inflammation, characterised by neutrophil and inflammatory monocyte recruitment and the production of inflammatory cytokines. We hypothesise that these changes to the lung environment alter the immune response and disease severity during subsequent RSV infection. To test this, we intranasally exposed mice to LPS, LTA or Acinetobacter baumannii (an airway bacterial pathogen) before RSV infection and observed an early induction of disease, measured by weight loss, at days 1-3 after infection. Neutrophils or inflammatory monocytes were not responsible for driving this exacerbated weight loss. Instead, exacerbated disease was associated with increased IL-1α and TNF-α, which orchestrated the recruitment of innate immune cells into the lung. This study shows that exposure to bacterial PAMPs prior to RSV infection increases the expression of IL-1α and TNF-α, which dysregulate the immune response resulting in exacerbated disease.
Collapse
Affiliation(s)
- Amber R Owen
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Ana Farias
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Anne-Marie Levins
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Sophie L Higham
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Cecilia Johansson
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom.
| |
Collapse
|
16
|
Dentice Maidana S, Argañaraz Aybar JN, Albarracin L, Imamura Y, Arellano-Arriagada L, Namai F, Suda Y, Nishiyama K, Villena J, Kitazawa H. Modulation of the Gut-Lung Axis by Water Kefir and Kefiran and Their Impact on Toll-like Receptor 3-Mediated Respiratory Immunity. Biomolecules 2024; 14:1457. [PMID: 39595633 PMCID: PMC11591811 DOI: 10.3390/biom14111457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The beneficial effect of milk kefir on respiratory heath has been previously demonstrated; however, water kefir and kefiran in the context of respiratory viral infections have not been investigated. Water kefir and kefiran could be alternatives to milk kefir for their application in persons with lactose intolerance or milk allergy and could be incorporated into vegan diets. Using mice models, this work demonstrated that the oral administration of water kefir or kefiran can modulate the respiratory Toll-like receptor (TLR3)-mediated innate antiviral immunity and improve the resistance to respiratory syncytial virus (RSV) infection. The treatment of mice with water kefir or kefiran for 6 days improved the production of interferons (IFN-β and IFN-γ) and antiviral factors (Mx2, OAS1, RNAseL, and IFITM3) in the respiratory tract after the activation of the TLR3 signaling pathway, differentially modulated the balance of pro- and anti-inflammatory cytokines, reduced RSV replication, and diminished lung tissue damage. Maintaining a proper balance between anti-inflammatory and pro-inflammatory mediators is vital for ensuring an effective and safe antiviral immune response, and the results of this work show that water kefir and kefiran would help to maintain that balance promoting a controlled inflammatory response that defends against infection while minimizing tissue damage.
Collapse
Affiliation(s)
- Stefania Dentice Maidana
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (S.D.M.); (L.A.)
| | - Julio Nicolás Argañaraz Aybar
- Cátedra de Inmunología, Instituto de Microbiología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán 4000, Argentina;
| | - Leonardo Albarracin
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (S.D.M.); (L.A.)
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (Y.I.); (L.A.-A.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Yoshiya Imamura
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (Y.I.); (L.A.-A.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Luciano Arellano-Arriagada
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (Y.I.); (L.A.-A.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Fu Namai
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (Y.I.); (L.A.-A.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Yoshihito Suda
- Department of Food, Agriculture and Environment, Miyagi University, Sendai 980-8572, Japan;
| | - Keita Nishiyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (Y.I.); (L.A.-A.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (S.D.M.); (L.A.)
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (Y.I.); (L.A.-A.); (F.N.); (K.N.)
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (Y.I.); (L.A.-A.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| |
Collapse
|
17
|
Gómez-Oro C, Latorre MC, Arribas-Poza P, Ibáñez-Escribano A, Baca-Cornejo KR, Gallego-Valle J, López-Escobar N, Mondéjar-Palencia M, Pion M, López-Fernández LA, Mercader E, Pérez-Milán F, Relloso M. Progesterone promotes CXCl2-dependent vaginal neutrophil killing by activating cervical resident macrophage-neutrophil crosstalk. JCI Insight 2024; 9:e177899. [PMID: 39298265 PMCID: PMC11529979 DOI: 10.1172/jci.insight.177899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Vaginal infections in women of reproductive age represent a clinical dilemma with significant socioeconomic implications. The current understanding of mucosal immunity failure during early pathogenic invasions that allows the pathogen to grow and thrive is far from complete. Neutrophils infiltrate most tissues following circadian patterns as part of normal repair, regulation of microbiota, or immune surveillance and become more numerous after infection. Neutrophils are responsible for maintaining vaginal immunity. Specific to the vagina, neutrophils continuously infiltrate at high levels, although during ovulation, they retreat to avoid sperm damage and permit reproduction. Here we show that, after ovulation, progesterone promotes resident vaginal macrophage-neutrophil crosstalk by upregulating Yolk sac early fetal organs (FOLR2+) macrophage CXCl2 expression, in a TNFA-patrolling monocyte-derived macrophage-mediated (CX3CR1hiMHCIIhi-mediated) manner, to activate the neutrophils' capacity to eliminate sex-transmitted and opportunistic microorganisms. Indeed, progesterone plays an essential role in conciliating the balance between the commensal microbiota, sperm, and the threat of pathogens because progesterone not only promotes a flurry of neutrophils but also increases neutrophilic fury to restore immunity after ovulation to thwart pathogenic invasion after intercourse. Therefore, modest progesterone dysregulations could lead to a suboptimal neutrophilic response, resulting in insufficient mucosal defense and recurrent unresolved infections.
Collapse
Affiliation(s)
- Carla Gómez-Oro
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Maria C. Latorre
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Patricia Arribas-Poza
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Alexandra Ibáñez-Escribano
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Katia R. Baca-Cornejo
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | | | - Natalia López-Escobar
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Mabel Mondéjar-Palencia
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marjorie Pion
- Laboratorio de InmunoRegulación, IiSGM, Madrid, Spain
| | - Luis A. López-Fernández
- Laboratorio de Farmacogenética, Grupo de Farmacia Hospitalaria y Farmacogenómica, IiSGM, Madrid, Spain
| | - Enrique Mercader
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Unidad Cirugía Endocrino-metabólica, Servicio de Cirugía General y Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Federico Pérez-Milán
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Unidad de Reproducción Asistida, Servicio de Obstetricia y Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Miguel Relloso
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
18
|
Martínez-Espinoza I, Babawale PI, Miletello H, Cheemarla NR, Guerrero-Plata A. Interferon Epsilon-Mediated Antiviral Activity Against Human Metapneumovirus and Respiratory Syncytial Virus. Vaccines (Basel) 2024; 12:1198. [PMID: 39460364 PMCID: PMC11511582 DOI: 10.3390/vaccines12101198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Interferon epsilon (IFN-ε) is a type I IFN that plays a critical role in the host immune response against pathogens. Despite having demonstrated antiviral activity in macrophages and mucosal tissues such as the female reproductive tract and the constitutive expression in mucosal tissues such as the lung, the relevance of IFN-ε against respiratory viral infections remains elusive. RESULTS We present, for the first time, the expression of IFN-ε in alveolar epithelial cells and primary human bronchial epithelial cells grown in an air-liquid interface (ALI) in response to human metapneumovirus (HMPV) and respiratory syncytial virus (RSV) infection. The molecular characterization of the IFN-ε induction by the viruses indicates that the expression of RIG-I is necessary for an optimal IFN-ε expression. Furthermore, treatment of the airway epithelial cells with rhIFN-ε induced the expression of IFN-stimulated genes (ISGs) and significantly restricted the viral replication of HMPV and RSV. CONCLUSIONS These findings underscore the relevance of IFN-ε against viral infections in the respiratory tract.
Collapse
Affiliation(s)
| | | | | | | | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
19
|
Anderson J, Do LAH, van Kasteren PB, Licciardi PV. The role of respiratory syncytial virus G protein in immune cell infection and pathogenesis. EBioMedicine 2024; 107:105318. [PMID: 39217853 PMCID: PMC11402919 DOI: 10.1016/j.ebiom.2024.105318] [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: 06/11/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
Severe respiratory syncytial virus (RSV) disease is a significant contributor to the global burden of disease in infants and children. The RSV attachment protein (G) has been shown to be critical in invading airway epithelial cells through its CX3C motif interacting with the host receptor CX3CR1. The ubiquitous expression of this receptor on immune cells may explain their susceptibility to RSV infection. The RSV G protein may enhance disease severity through reprogramming of normal cellular functionality leading to inhibition of antiviral responses. While existing preventives targeting the RSV fusion (F) protein are highly effective, there are no RSV therapeutics based on the G protein to limit RSV pathogenesis. Monoclonal antibodies targeting the RSV G protein administered as post-infection therapeutics in mice have been shown to improve the antiviral response, reduce viral load and limit disease severity. Further research is required to better understand how RSV infection of immune cells contributes to pathogenesis for the development of more targeted and efficacious therapeutics.
Collapse
Affiliation(s)
- Jeremy Anderson
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - Lien Anh Ha Do
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Puck B van Kasteren
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Paul V Licciardi
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
20
|
Lum KK, Reed TJ, Yang J, Cristea IM. Differential Contributions of Interferon Classes to Host Inflammatory Responses and Restricting Virus Progeny Production. J Proteome Res 2024; 23:3249-3268. [PMID: 38564653 PMCID: PMC11296908 DOI: 10.1021/acs.jproteome.3c00826] [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: 04/04/2024]
Abstract
Fundamental to mammalian intrinsic and innate immune defenses against pathogens is the production of Type I and Type II interferons, such as IFN-β and IFN-γ, respectively. The comparative effects of IFN classes on the cellular proteome, protein interactions, and virus restriction within cell types that differentially contribute to immune defenses are needed for understanding immune signaling. Here, a multilayered proteomic analysis, paired with biochemical and molecular virology assays, allows distinguishing host responses to IFN-β and IFN-γ and associated antiviral impacts during infection with several ubiquitous human viruses. In differentiated macrophage-like monocytic cells, we classified proteins upregulated by IFN-β, IFN-γ, or pro-inflammatory LPS. Using parallel reaction monitoring, we developed a proteotypic peptide library for shared and unique ISG signatures of each IFN class, enabling orthogonal confirmation of protein alterations. Thermal proximity coaggregation analysis identified the assembly and maintenance of IFN-induced protein interactions. Comparative proteomics and cytokine responses in macrophage-like monocytic cells and primary keratinocytes provided contextualization of their relative capacities to restrict virus production during infection with herpes simplex virus type-1, adenovirus, and human cytomegalovirus. Our findings demonstrate how IFN classes induce distinct ISG abundance and interaction profiles that drive antiviral defenses within cell types that differentially coordinate mammalian immune responses.
Collapse
Affiliation(s)
- Krystal K. Lum
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544
| | - Tavis J. Reed
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544
| | - Jinhang Yang
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544
| |
Collapse
|
21
|
Burgess MO, Janas P, Berry K, Mayr H, Mack M, Jenkins SJ, Bain CC, McSorley HJ, Schwarze J. Helminth induced monocytosis conveys protection from respiratory syncytial virus infection in mice. Allergy 2024; 79:2157-2172. [PMID: 38924546 DOI: 10.1111/all.16206] [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: 05/10/2023] [Revised: 04/17/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection in infants is a major cause of viral bronchiolitis and hospitalisation. We have previously shown in a murine model that ongoing infection with the gut helminth Heligmosomoides polygyrus protects against RSV infection through type I interferon (IFN-I) dependent reduction of viral load. Yet, the cellular basis for this protection has remained elusive. Given that recruitment of mononuclear phagocytes to the lung is critical for early RSV infection control, we assessed their role in this coinfection model. METHODS Mice were infected by oral gavage with H. polygyrus. Myeloid immune cell populations were assessed by flow cytometry in lung, blood and bone marrow throughout infection and after secondary infection with RSV. Monocyte numbers were depleted by anti-CCR2 antibody or increased by intravenous transfer of enriched monocytes. RESULTS H. polygyrus infection induces bone marrow monopoiesis, increasing circulatory monocytes and lung mononuclear phagocytes in a IFN-I signalling dependent manner. This expansion causes enhanced lung mononuclear phagocyte counts early in RSV infection that may contribute to the reduction of RSV load. Depletion or supplementation of circulatory monocytes prior to RSV infection confirms that these are both necessary and sufficient for helminth induced antiviral protection. CONCLUSIONS H. polygyrus infection induces systemic monocytosis contributing to elevated mononuclear phagocyte numbers in the lung. These cells are central to an anti-viral effect that reduces the peak viral load in RSV infection. Treatments to promote or modulate these cells may provide novel paths to control RSV infection in high risk individuals.
Collapse
Affiliation(s)
- Matthew O Burgess
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Piotr Janas
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Karla Berry
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Hannah Mayr
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Stephen J Jenkins
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Calum C Bain
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Henry J McSorley
- Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Jurgen Schwarze
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Child Life and Health, Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
22
|
Farazuddin M, Acker G, Zourob J, O’Konek JJ, Wong PT, Morris S, Rasky AJ, Kim CH, Lukacs NW, Baker JR. Inhibiting retinoic acid signaling in dendritic cells suppresses respiratory syncytial virus infection through enhanced antiviral immunity. iScience 2024; 27:110103. [PMID: 39045100 PMCID: PMC11263793 DOI: 10.1016/j.isci.2024.110103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/21/2023] [Accepted: 05/21/2024] [Indexed: 07/25/2024] Open
Abstract
Retinoic acid (RA), controls the immunoregulatory functions of many immune cells, including dendritic cells (DCs), and is important for mucosal immunity. In DCs, RA regulates the expression of pattern recognition receptors and stimulates interferon production. Here, we investigated the role of RA in DCs in mounting immunity to respiratory syncytial virus (RSV). To abolish RA signaling in DCs, we used mice expressing a dominant negative form of retinoic acid receptor-α (RARα) under the CD11c promoter (CD11c-dnRARα). Paradoxically, upon RSV challenge, these animals had lower viral burden, reduced pathology, and greater Th1 polarized immunity than wild-type (WT) mice. Moreover, CD11c-dnRARα DCs infected with RSV showed enhancement in innate and adaptive immunity genes, while genes associated with viral replication were downregulated. These findings suggest that the absence of RA signaling in DCs enhances innate immunity against RSV infection leading to decreased viral load and reduced pathogenicity.
Collapse
Affiliation(s)
- Mohammad Farazuddin
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Grant Acker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Joseph Zourob
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jessica J. O’Konek
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pamela T. Wong
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Susan Morris
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew J. Rasky
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Chang H. Kim
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James R. Baker
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
- Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Ogonczyk-Makowska D, Brun P, Vacher C, Chupin C, Droillard C, Carbonneau J, Laurent E, Dulière V, Traversier A, Terrier O, Julien T, Galloux M, Paul S, Eléouët JF, Fouret J, Hamelin ME, Pizzorno A, Boivin G, Rosa-Calatrava M, Dubois J. Mucosal bivalent live attenuated vaccine protects against human metapneumovirus and respiratory syncytial virus in mice. NPJ Vaccines 2024; 9:111. [PMID: 38898106 PMCID: PMC11187144 DOI: 10.1038/s41541-024-00899-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Live-Attenuated Vaccines (LAVs) stimulate robust mucosal and cellular responses and have the potential to protect against Respiratory Syncytial Virus (RSV) and Human Metapneumovirus (HMPV), the main etiologic agents of viral bronchiolitis and pneumonia in children. We inserted the RSV-F gene into an HMPV-based LAV (Metavac®) we previously validated for the protection of mice against HMPV challenge, and rescued a replicative recombinant virus (Metavac®-RSV), exposing both RSV- and HMPV-F proteins at the virion surface and expressing them in reconstructed human airway epithelium models. When administered to BALB/c mice by the intranasal route, bivalent Metavac®-RSV demonstrated its capacity to replicate with reduced lung inflammatory score and to protect against both RSV and lethal HMPV challenges in vaccinated mice while inducing strong IgG and broad RSV and HMPV neutralizing antibody responses. Altogether, our results showed the versatility of the Metavac® platform and suggested that Metavac®-RSV is a promising mucosal bivalent LAV candidate to prevent pneumovirus-induced diseases.
Collapse
Affiliation(s)
- Daniela Ogonczyk-Makowska
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Québec, QC, G1V 4G2, Canada
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
| | - Pauline Brun
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Clémence Vacher
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Québec, QC, G1V 4G2, Canada
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Caroline Chupin
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Vaxxel, 43 Boulevard du onze novembre 1918, 69100, Villeurbanne, France
| | - Clément Droillard
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Julie Carbonneau
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Québec, QC, G1V 4G2, Canada
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
| | - Emilie Laurent
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Victoria Dulière
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Aurélien Traversier
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Thomas Julien
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Marie Galloux
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Stéphane Paul
- CIRI, Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, INSERM U1111, CNRS UMR5308, ENS Lyon, Université Jean Monnet Saint-Etienne, Saint-Etienne, France
| | | | - Julien Fouret
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Nexomis, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Marie-Eve Hamelin
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Québec, QC, G1V 4G2, Canada
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
| | - Andrés Pizzorno
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Guy Boivin
- Centre de Recherche en Infectiologie of the Centre Hospitalier Universitaire de Québec and Université Laval, Québec, QC, G1V 4G2, Canada
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
| | - Manuel Rosa-Calatrava
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France
| | - Julia Dubois
- International Research Laboratory RESPIVIR France - Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec, 69008, Lyon, France, Université Claude Bernard Lyon 1, Université de Lyon, INSERM, CNRS, ENS de Lyon, France, Centre Hospitalier Universitaire de Québec - Université Laval, QC G1V 4G2, Québec, Canada.
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, INSERM U1111, CNRS UMR 5308, ENS de Lyon, Université Claude Bernard Lyon 1, Lyon, France.
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, 69008, Lyon, France.
| |
Collapse
|
24
|
Gambadauro A, Galletta F, Li Pomi A, Manti S, Piedimonte G. Immune Response to Respiratory Viral Infections. Int J Mol Sci 2024; 25:6178. [PMID: 38892370 PMCID: PMC11172738 DOI: 10.3390/ijms25116178] [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: 05/07/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The respiratory system is constantly exposed to viral infections that are responsible for mild to severe diseases. In this narrative review, we focalized the attention on respiratory syncytial virus (RSV), influenza virus, and severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infections, responsible for high morbidity and mortality in the last decades. We reviewed the human innate and adaptive immune responses in the airways following infection, focusing on a particular population: newborns and pregnant women. The recent Coronavirus disease-2019 (COVID-19) pandemic has highlighted how our interest in viral pathologies must not decrease. Furthermore, we must increase our knowledge of infection mechanisms to improve our future defense strategies.
Collapse
Affiliation(s)
- Antonella Gambadauro
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Francesca Galletta
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Alessandra Li Pomi
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Sara Manti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (A.G.); (F.G.); (A.L.P.)
| | - Giovanni Piedimonte
- Office for Research and Departments of Pediatrics, Biochemistry, and Molecular Biology, Tulane University, New Orleans, LA 70112, USA;
| |
Collapse
|
25
|
Pernet E, Poschmann J, Divangahi M. A complex immune communication between eicosanoids and pulmonary macrophages. Curr Opin Virol 2024; 66:101399. [PMID: 38547562 DOI: 10.1016/j.coviro.2024.101399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 06/07/2024]
Abstract
Respiratory viral infections represent a constant threat for human health and urge for a better understanding of the pulmonary immune response to prevent disease severity. Macrophages are at the center of pulmonary immunity, where they play a pivotal role in orchestrating beneficial and/or pathological outcomes during infection. Eicosanoids, the host bioactive lipid mediators, have re-emerged as important regulators of pulmonary immunity during respiratory viral infections. In this review, we summarize the current knowledge linking eicosanoids' and pulmonary macrophages' homeostatic and antimicrobial functions and discuss eicosanoids as emerging targets for immunotherapy in viral infection.
Collapse
Affiliation(s)
- Erwan Pernet
- Department of Medical Biology, Université du Québec à Trois-Rivières, Québec, Canada.
| | - Jeremie Poschmann
- INSERM, Nantes Université, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Maziar Divangahi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| |
Collapse
|
26
|
Lim PN, Cervantes MM, Pham LK, Doherty S, Tufts A, Dubey D, Mai D, Aderem A, Diercks AH, Rothchild AC. Absence of c-Maf and IL-10 enables Type I IFN enhancement of innate responses to low-dose LPS in alveolar macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.594428. [PMID: 38826239 PMCID: PMC11142172 DOI: 10.1101/2024.05.22.594428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Alveolar macrophages (AMs) are lower-airway resident myeloid cells and are among the first to respond to inhaled pathogens. Here, we interrogate AM innate sensing to Pathogen Associated Molecular Patterns (PAMPs) and determine AMs have decreased responses to low-dose LPS compared to other macrophages, as measured by TNF, IL-6, Ifnb, and Ifit3. We find the reduced response to low-dose LPS correlates with minimal TLR4 and CD14 surface expression, despite sufficient internal expression of TLR4. Additionally, we find that AMs do not produce IL-10 in response to a variety of PAMPs due to low expression of transcription factor c-Maf and that lack of IL-10 production contributes to an enhancement of pro-inflammatory responses by Type I IFN. Our findings demonstrate that AMs have cell-intrinsic dampened responses to LPS, which is enhanced by type I IFN exposure. These data implicate conditions where AMs may have reduced or enhanced sentinel responses to bacterial infections.
Collapse
Affiliation(s)
- Pamelia N. Lim
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Maritza M. Cervantes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Linh K. Pham
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Graduate Program in Animal Biotechnology & Biomedical Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Sydney Doherty
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Ankita Tufts
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Divya Dubey
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98019
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98019
| | - Alan H. Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98019
| | - Alissa C. Rothchild
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
27
|
Parsons EL, Kim JS, Malloy AMW. Development of innate and adaptive immunity to RSV in young children. Cell Immunol 2024; 399-400:104824. [PMID: 38615612 DOI: 10.1016/j.cellimm.2024.104824] [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: 12/24/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
Infection of the respiratory tract with respiratory syncytial virus (RSV) is common and occurs repeatedly throughout life with most severe disease occurring at the extremes of age: in young infants and the elderly. Effective anti-viral therapeutics are not available and therefore prevention has been the primary strategy for reducing the disease burden. Our current understanding of respiratory mucosal cell biology and the immune response within the respiratory tract is inadequate to prevent infection caused by a pathogen like RSV that does not disseminate outside of this environment. Gaps in our understanding of the activation of innate and adaptive immunity in response to RSV and the role of age upon infection also limit improvements in the design of therapeutics and vaccines for young infants. However, advancements in structural biology have improved our ability to characterize antibodies against viral proteins and in 2023 the first vaccines for those over 60 years and pregnant women became available, potentially reducing the burden of disease. This review will examine our current understanding of the critical facets of anti-RSV immune responses in infants and young children as well as highlight areas where more research is needed.
Collapse
Affiliation(s)
| | - Jisung S Kim
- Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation, Bethesda, MD, USA
| | | |
Collapse
|
28
|
Anderson J, Imran S, Ng YY, Wang T, Ashley S, Minh Thang C, Quang Thanh L, Thi Trang Dai V, Van Thanh P, Thi Hong Nhu B, Ngoc Xuan Trang D, Thi Phuong Trinh P, Thanh Binh L, Thuong Vu N, Trong Toan N, Novakovic B, Tang MLK, Wurzel D, Mulholland K, Pellicci DG, Do LAH, Licciardi PV. Differential anti-viral response to respiratory syncytial virus A in preterm and term infants. EBioMedicine 2024; 102:105044. [PMID: 38447274 PMCID: PMC10933467 DOI: 10.1016/j.ebiom.2024.105044] [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: 09/26/2023] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Preterm infants are more likely to experience severe respiratory syncytial virus (RSV) disease compared to term infants. The reasons for this are multi-factorial, however their immature immune system is believed to be a major contributing factor. METHODS We collected cord blood from 25 preterm (gestational age 30.4-34.1 weeks) and 25 term infants (gestation age 37-40 weeks) and compared the response of cord blood mononuclear cells (CBMCs) to RSVA and RSVB stimulation using neutralising assays, high-dimensional flow cytometry, multiplex cytokine assays and RNA-sequencing. FINDINGS We found that preterm and term infants had similar maternally derived neutralising antibody titres to RSVA and RSVB. Preterm infants had significantly higher myeloid dendritic cells (mDC) RSV infection compared to term infants. Differential gene expression analysis of RSVA stimulated CBMCs revealed enrichment of genes involved in cytokine production and immune regulatory pathways involving IL-10, IL-36γ, CXCL1, CXCL2, SOCS1 and SOCS3 in term infants, while differentially expressed genes (DEGs) in preterm infants were related to cell cycle (CDK1, TTK, ESCO2, KNL1, CDC25A, MAD2L1) without associated expression of immune response genes. Furthermore, enriched genes in term infants were highly correlated suggesting an increased co-ordination of their immune response to RSVA. When comparing DEGs in preterm and term infants following RSVB stimulation, no differences in immune response genes were identified. INTERPRETATION Overall, our data suggests that preterm infants have a more restricted immunological response to RSVA compared with term infants. While further studies are required, these findings may help to explain why preterm infants are more susceptible to severe RSV disease and identify potential therapeutic targets to protect these vulnerable infants. FUNDING Murdoch Children's Research Institute Infection and Immunity theme grant.
Collapse
Affiliation(s)
- Jeremy Anderson
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - Samira Imran
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Yan Yung Ng
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Tongtong Wang
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia
| | - Sarah Ashley
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | - Boris Novakovic
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Mimi L K Tang
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia; Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia
| | - Danielle Wurzel
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia; Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia; Royal Children's Hospital, Melbourne, Australia
| | - Kim Mulholland
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia; London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Daniel G Pellicci
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
| | - Lien Anh Ha Do
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - Paul V Licciardi
- Infection, Immunity and Global Health, Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
29
|
Majumder S, Das S, Li P, Yang N, Dellario H, Sui H, Guan Z, Sun W. Pneumonic Plague Protection Induced by a Monophosphoryl Lipid A Decorated Yersinia Outer-Membrane-Vesicle Vaccine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307066. [PMID: 38009518 PMCID: PMC11009084 DOI: 10.1002/smll.202307066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/19/2023] [Indexed: 11/29/2023]
Abstract
A new Yersinia pseudotuberculosis mutant strain, YptbS46, carrying the lpxE insertion and pmrF-J deletion is constructed and shown to exclusively produce monophosphoryl lipid A (MPLA) having adjuvant properties. Outer membrane vesicles (OMVs) isolated from YptbS46 harboring an lcrV expression plasmid, pSMV13, are designated OMV46-LcrV, which contained MPLA and high amounts of LcrV (Low Calcium response V) and displayed low activation of Toll-like receptor 4 (TLR4). Intramuscular prime-boost immunization with 30 µg of of OMV46-LcrV exhibited substantially reduced reactogenicity than the parent OMV44-LcrV and conferred complete protection to mice against a high-dose of respiratory Y. pestis challenge. OMV46-LcrV immunization induced robust adaptive responses in both lung mucosal and systemic compartments and orchestrated innate immunity in the lung, which are correlated with rapid bacterial clearance and unremarkable lung damage during Y. pestis challenge. Additionally, OMV46-LcrV immunization conferred long-term protection. Moreover, immunization with reduced doses of OMV46-LcrV exhibited further lower reactogenicity and still provided great protection against pneumonic plague. The studies strongly demonstrate the feasibility of OMV46-LcrV as a new type of plague vaccine candidate.
Collapse
Affiliation(s)
- Saugata Majumder
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Shreya Das
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Peng Li
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Nicole Yang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Hazel Dellario
- Wadsworth Center, New York State Department of Health, Albany, NY, 12237, USA
| | - Haixin Sui
- Wadsworth Center, New York State Department of Health, Albany, NY, 12237, USA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, USA
| | - Wei Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| |
Collapse
|
30
|
Baker PJ, Bohrer AC, Castro E, Amaral EP, Snow-Smith M, Torres-Juárez F, Gould ST, Queiroz ATL, Fukutani ER, Jordan CM, Khillan JS, Cho K, Barber DL, Andrade BB, Johnson RF, Hilligan KL, Mayer-Barber KD. The inflammatory microenvironment of the lung at the time of infection governs innate control of SARS-CoV-2 replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.586885. [PMID: 38585846 PMCID: PMC10996686 DOI: 10.1101/2024.03.27.586885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
SARS-CoV-2 infection leads to vastly divergent clinical outcomes ranging from asymptomatic infection to fatal disease. Co-morbidities, sex, age, host genetics and vaccine status are known to affect disease severity. Yet, how the inflammatory milieu of the lung at the time of SARS-CoV-2 exposure impacts the control of viral replication remains poorly understood. We demonstrate here that immune events in the mouse lung closely preceding SARS-CoV-2 infection significantly impact viral control and we identify key innate immune pathways required to limit viral replication. A diverse set of pulmonary inflammatory stimuli, including resolved antecedent respiratory infections with S. aureus or influenza, ongoing pulmonary M. tuberculosis infection, ovalbumin/alum-induced asthma or airway administration of defined TLR ligands and recombinant cytokines, all establish an antiviral state in the lung that restricts SARS-CoV-2 replication upon infection. In addition to antiviral type I interferons, the broadly inducible inflammatory cytokines TNFα and IL-1 precondition the lung for enhanced viral control. Collectively, our work shows that SARS-CoV-2 may benefit from an immunologically quiescent lung microenvironment and suggests that heterogeneity in pulmonary inflammation that precedes or accompanies SARS-CoV-2 exposure may be a significant factor contributing to the population-wide variability in COVID-19 disease outcomes.
Collapse
Affiliation(s)
- Paul J. Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Current Address: Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Andrea C. Bohrer
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Ehydel Castro
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Eduardo P. Amaral
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Maryonne Snow-Smith
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
- Human Eosinophil Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Flor Torres-Juárez
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Sydnee T. Gould
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
- Current Address: Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Artur T. L. Queiroz
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Eduardo R. Fukutani
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Cassandra M. Jordan
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Jaspal S. Khillan
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Kyoungin Cho
- Mouse Genetics and Gene Modification Section, Comparative Medicine Branch, NIAID, NIH, Rockville, Maryland 20852, USA
| | - Daniel L. Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Bruno B. Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, Salvador, Bahia 41810-710, Brazil
- Laboratory of Clinical and Translational Research, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia 40296-710, Brazil
| | - Reed F. Johnson
- SCV2 Virology Core, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland 20892, USA
| | - Kerry L. Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| |
Collapse
|
31
|
Chappin K, Besteman SB, Hennus MP, Wildenbeest JG, Mokry M, Bont LJ, van der Vlist M, Calis JJA. Airway and Blood Monocyte Transcriptomic Profiling Reveals an Antiviral Phenotype in Infants With Severe Respiratory Syncytial Virus Infection. J Infect Dis 2024; 229:S100-S111. [PMID: 37941411 DOI: 10.1093/infdis/jiad487] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection is the primary cause of lower respiratory tract infections in children <5 years of age. Monocytes, especially in the respiratory tract, are suggested to contribute to RSV pathology, but their role is incompletely understood. With transcriptomic profiling of blood and airway monocytes, we describe the role of monocytes in severe RSV infection. METHODS Tracheobronchial aspirates and blood samples were collected from control patients (n = 9) and those infected with RSV (n = 14) who were admitted to the pediatric intensive care unit. Monocytes (CD14+) were sorted and analyzed by RNA sequencing for transcriptomic profiling. RESULTS Peripheral blood and airway monocytes of patients with RSV demonstrated increased expression of antiviral and interferon-responsive genes as compared with controls. Cytokine signaling showed a shared response between blood and airway monocytes while displaying responses that were more pronounced according to the tissue of origin. Airway monocytes upregulated additional genes related to migration and inflammation. CONCLUSIONS We found that the RSV-induced interferon response extends from the airways to the peripheral blood. Moreover, RSV induces a migration-promoting transcriptional program in monocytes. Unraveling the monocytic response and its role in the immune response to RSV infection could help the development of therapeutics to prevent severe disease.
Collapse
Affiliation(s)
- K Chappin
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University
| | | | - M P Hennus
- Department of Paediatric Intensive Care, Wilhelmina Children's Hospital, University Medical Centre Utrecht
| | | | - M Mokry
- Experimental Cardiology, Department of Heart and Lungs, University Medical Centre Utrecht, the Netherlands
| | - L J Bont
- Department of Paediatric Infectious Diseases and Immunology
| | - M van der Vlist
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University
| | - J J A Calis
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University
| |
Collapse
|
32
|
Li W, Wang X, Chen Y, Ding Y, Ling X, Yuan B, Tao J. Luteolin-7-O-glucoside promotes macrophage release of IFN-β by maintaining mitochondrial function and corrects the disorder of glucose metabolism during RSV infection. Eur J Pharmacol 2024; 963:176271. [PMID: 38113965 DOI: 10.1016/j.ejphar.2023.176271] [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: 07/11/2023] [Revised: 11/26/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
Respiratory syncytial virus (RSV) pneumonia is the main cause of acute bronchiolitis in infants. Luteolin-7-O-glucoside (LUT-7G) is a natural flavonoid, which exists in a variety of plants and has the potential to treat viral pneumonia. We established RSV pneumonia mouse models and RSV-infected cell models. Clodronate liposomes were used to deplete macrophages. We used HE staining and immunohistochemistry to determine inflammatory damage and virus replication. We detected the expression levels of inflammatory factors and IFN-β through qPCR and ELISA. JC-1 kit was used for detecting the cell mitochondrial Membrane potential (MMP). ROS, SOD, and MDA kits were used for detecting intracellular oxidative stress damage. Metabolites of TCA in lung tissue and serum of mice were detected by GC-MS. Pharmacodynamic studies have shown that intervention with LUT-7G can alleviate lung tissue damage caused by RSV infection, inhibit RSV replication, and downregulate TNF-α, IL-1β, and IL-6 mRNA expression. LUT-7G upregulated the IFN-β content and the expression of IFN-β, ISG15, and OAS1 mRNA. In vitro, LUT-7G inhibited RSV-induced cell death, reversed the RSV-induced decrease of MMP and decreased intracellular oxidative stress. Target metabonomics showed that RSV infection upregulated the levels of glycolysis and TCA metabolites in lung tissue and serum, while LUT-7G could improve the disorder of glucose metabolism. The results indicate that LUT-7G can promote the release of IFN-β in the lung, alleviate inflammatory damage, and inhibit RSV replication during RSV infection. These effects may be achieved by protecting the mitochondrial function of alveolar macrophages and correcting the disorder of glucose metabolism.
Collapse
Affiliation(s)
- Weifeng Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China; Jiangsu Key Laboratory of Paediatric Respiratory Disease, Institute of Paediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xuan Wang
- Jiangsu Vocational College of Medicine, Yancheng, 224000, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yanzhen Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Oncology Department, Nanjing, 210023, China.
| | - Yali Ding
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China; Jiangsu Key Laboratory of Paediatric Respiratory Disease, Institute of Paediatrics, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaoyin Ling
- Affiliated Hospital of Nantong University, Nantong, 226000, China.
| | - Bin Yuan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China.
| | - Jialei Tao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Department of Paediatrics, Nanjing, 210023, China.
| |
Collapse
|
33
|
Makdissi N. Macrophage Development and Function. Methods Mol Biol 2024; 2713:1-9. [PMID: 37639112 DOI: 10.1007/978-1-0716-3437-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Macrophages were first described over a hundred years ago. Throughout the years, they were shown to be essential players in their tissue-specific environment, performing various functions during homeostatic and disease conditions. Recent reports shed more light on their ontogeny as long-lived, self-maintained cells with embryonic origin in most tissues. They populate the different tissues early during development, where they help to establish and maintain homeostasis. In this chapter, the history of macrophages is discussed. Furthermore, macrophage ontogeny and core functions in the different tissues are described.
Collapse
Affiliation(s)
- Nikola Makdissi
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
34
|
Drake KA, Talantov D, Tong GJ, Lin JT, Verheijden S, Katz S, Leung JM, Yuen B, Krishna V, Wu MJ, Sutherland AM, Short SA, Kheradpour P, Mumbach MR, Franz KM, Trifonov V, Lucas MV, Merson J, Kim CC. Multi-omic profiling reveals early immunological indicators for identifying COVID-19 Progressors. Clin Immunol 2023; 256:109808. [PMID: 37852344 DOI: 10.1016/j.clim.2023.109808] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/25/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
We sought to better understand the immune response during the immediate post-diagnosis phase of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by identifying molecular associations with longitudinal disease outcomes. Multi-omic analyses identified differences in immune cell composition, cytokine levels, and cell subset-specific transcriptomic and epigenomic signatures between individuals on a more serious disease trajectory (Progressors) as compared to those on a milder course (Non-progressors). Higher levels of multiple cytokines were observed in Progressors, with IL-6 showing the largest difference. Blood monocyte cell subsets were also skewed, showing a comparative decrease in non-classical CD14-CD16+ and intermediate CD14+CD16+ monocytes. In lymphocytes, the CD8+ T effector memory cells displayed a gene expression signature consistent with stronger T cell activation in Progressors. These early stage observations could serve as the basis for the development of prognostic biomarkers of disease risk and interventional strategies to improve the management of severe COVID-19. BACKGROUND: Much of the literature on immune response post-SARS-CoV-2 infection has been in the acute and post-acute phases of infection. TRANSLATIONAL SIGNIFICANCE: We found differences at early time points of infection in approximately 160 participants. We compared multi-omic signatures in immune cells between individuals progressing to needing more significant medical intervention and non-progressors. We observed widespread evidence of a state of increased inflammation associated with progression, supported by a range of epigenomic, transcriptomic, and proteomic signatures. The signatures we identified support other findings at later time points and serve as the basis for prognostic biomarker development or to inform interventional strategies.
Collapse
Affiliation(s)
- Katherine A Drake
- Verily Life Sciences, South San Francisco, CA, United States of America
| | - Dimitri Talantov
- Janssen Research & Development, LLC, San Diego, CA, United States of America
| | - Gary J Tong
- Verily Life Sciences, South San Francisco, CA, United States of America
| | - Jack T Lin
- Verily Life Sciences, South San Francisco, CA, United States of America
| | | | - Samuel Katz
- Verily Life Sciences, South San Francisco, CA, United States of America
| | | | - Benjamin Yuen
- Verily Life Sciences, South San Francisco, CA, United States of America
| | - Vinod Krishna
- Janssen Research & Development, LLC, San Diego, CA, United States of America
| | - Michelle J Wu
- Verily Life Sciences, South San Francisco, CA, United States of America
| | | | - Sarah A Short
- Verily Life Sciences, South San Francisco, CA, United States of America
| | - Pouya Kheradpour
- Verily Life Sciences, South San Francisco, CA, United States of America
| | - Maxwell R Mumbach
- Verily Life Sciences, South San Francisco, CA, United States of America
| | - Kate M Franz
- Verily Life Sciences, South San Francisco, CA, United States of America
| | - Vladimir Trifonov
- Janssen Research & Development, LLC, San Diego, CA, United States of America
| | - Molly V Lucas
- Janssen Research & Development, LLC, NJ, United States of America
| | - James Merson
- Janssen Research & Development, LLC, San Francisco, CA, United States of America
| | - Charles C Kim
- Verily Life Sciences, South San Francisco, CA, United States of America.
| |
Collapse
|
35
|
Almond M, Farne HA, Jackson MM, Jha A, Katsoulis O, Pitts O, Tunstall T, Regis E, Dunning J, Byrne AJ, Mallia P, Kon OM, Saunders KA, Simpson KD, Snelgrove RJ, Openshaw PJM, Edwards MR, Barclay WS, Heaney LM, Johnston SL, Singanayagam A. Obesity dysregulates the pulmonary antiviral immune response. Nat Commun 2023; 14:6607. [PMID: 37857661 PMCID: PMC10587167 DOI: 10.1038/s41467-023-42432-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Obesity is a well-recognized risk factor for severe influenza infections but the mechanisms underlying susceptibility are poorly understood. Here, we identify that obese individuals have deficient pulmonary antiviral immune responses in bronchoalveolar lavage cells but not in bronchial epithelial cells or peripheral blood dendritic cells. We show that the obese human airway metabolome is perturbed with associated increases in the airway concentrations of the adipokine leptin which correlated negatively with the magnitude of ex vivo antiviral responses. Exogenous pulmonary leptin administration in mice directly impaired antiviral type I interferon responses in vivo and ex vivo in cultured airway macrophages. Obese individuals hospitalised with influenza showed dysregulated upper airway immune responses. These studies provide insight into mechanisms driving propensity to severe influenza infections in obesity and raise the potential for development of leptin manipulation or interferon administration as novel strategies for conferring protection from severe infections in obese higher risk individuals.
Collapse
Affiliation(s)
- Mark Almond
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Hugo A Farne
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Millie M Jackson
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK
| | - Akhilesh Jha
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Orestis Katsoulis
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK
| | - Oliver Pitts
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK
| | | | - Eteri Regis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jake Dunning
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, UK
- School of Medicine and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, 4, Ireland
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Onn Min Kon
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | | | | | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Wendy S Barclay
- Section of Virology, Department of Infectious Disease, Imperial College London, London, UK
| | - Liam M Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | | | - Aran Singanayagam
- Centre for Bacterial Resistance Biology. Section of Molecular Microbiology. Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
36
|
Agac A, Kolbe SM, Ludlow M, Osterhaus ADME, Meineke R, Rimmelzwaan GF. Host Responses to Respiratory Syncytial Virus Infection. Viruses 2023; 15:1999. [PMID: 37896776 PMCID: PMC10611157 DOI: 10.3390/v15101999] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/29/2023] Open
Abstract
Respiratory syncytial virus (RSV) infections are a constant public health problem, especially in infants and older adults. Virtually all children will have been infected with RSV by the age of two, and reinfections are common throughout life. Since antigenic variation, which is frequently observed among other respiratory viruses such as SARS-CoV-2 or influenza viruses, can only be observed for RSV to a limited extent, reinfections may result from short-term or incomplete immunity. After decades of research, two RSV vaccines were approved to prevent lower respiratory tract infections in older adults. Recently, the FDA approved a vaccine for active vaccination of pregnant women to prevent severe RSV disease in infants during their first RSV season. This review focuses on the host response to RSV infections mediated by epithelial cells as the first physical barrier, followed by responses of the innate and adaptive immune systems. We address possible RSV-mediated immunomodulatory and pathogenic mechanisms during infections and discuss the current vaccine candidates and alternative treatment options.
Collapse
Affiliation(s)
| | | | | | | | | | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (A.A.); (S.M.K.); (M.L.); (A.D.M.E.O.); (R.M.)
| |
Collapse
|
37
|
da Silva RP, Thomé BL, da Souza APD. Exploring the Immune Response against RSV and SARS-CoV-2 Infection in Children. BIOLOGY 2023; 12:1223. [PMID: 37759622 PMCID: PMC10525162 DOI: 10.3390/biology12091223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
Viral respiratory tract infections are a significant public health concern, particularly in children. RSV is a prominent cause of lower respiratory tract infections among infants, whereas SARS-CoV-2 has caused a global pandemic with lower overall severity in children than in adults. In this review, we aimed to compare the innate and adaptive immune responses induced by RSV and SARS-CoV-2 to better understand differences in the pathogenesis of infection. Some studies have demonstrated that children present a more robust immune response against SARS-CoV-2 than adults; however, this response is dissimilar to that of RSV. Each virus has a distinctive mechanism to escape the immune response. Understanding the mechanisms underlying these differences is crucial for developing effective treatments and improving the management of pediatric respiratory infections.
Collapse
Affiliation(s)
| | | | - Ana Paula Duarte da Souza
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619-900, Brazil; (R.P.d.S.); (B.L.T.)
| |
Collapse
|
38
|
Bergeron HC, Hansen MR, Tripp RA. Interferons-Implications in the Immune Response to Respiratory Viruses. Microorganisms 2023; 11:2179. [PMID: 37764023 PMCID: PMC10535750 DOI: 10.3390/microorganisms11092179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Interferons (IFN) are an assemblage of signaling proteins made and released by various host cells in response to stimuli, including viruses. Respiratory syncytial virus (RSV), influenza virus, and SARS-CoV-2 are major causes of respiratory disease that induce or antagonize IFN responses depending on various factors. In this review, the role and function of type I, II, and III IFN responses to respiratory virus infections are considered. In addition, the role of the viral proteins in modifying anti-viral immunity is noted, as are the specific IFN responses that underly the correlates of immunity and protection from disease.
Collapse
Affiliation(s)
| | | | - Ralph A. Tripp
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA 30605, USA; (H.C.B.); (M.R.H.)
| |
Collapse
|
39
|
Majumder S, Das S, Li P, Yang N, Dellario H, Sui H, Guan Z, Sun W. Pneumonic plague protection induced by a monophosphoryl lipid A decorated Yersinia outer-membrane-vesicle vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553697. [PMID: 37645871 PMCID: PMC10462118 DOI: 10.1101/2023.08.17.553697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
A newly constructed Yersinia pseudotuberculosis mutant (YptbS46) carrying the lpxE insertion and pmrF-J deletion exclusively synthesized an adjuvant form of lipid A, monophosphoryl lipid A (MPLA). Outer membrane vesicles (OMVs) isolated from YptbS46 harboring an lcrV expression plasmid, pSMV13, were designated OMV 46 -LcrV, which contained MPLA and high amounts of LcrV and displayed low activation of Toll-like receptor 4 (TLR4). Similar to the previous OMV 44 -LcrV, intramuscular prime-boost immunization with 30 µg of OMV 46 -LcrV exhibited substantially reduced reactogenicity and conferred complete protection to mice against a high-dose of respiratory Y. pestis challenge. OMV 46 -LcrV immunization induced robust adaptive responses in both lung mucosal and systemic compartments and orchestrated innate immunity in the lung, which were correlated with rapid bacterial clearance and unremarkable lung damage during Y. pestis challenge. Additionally, OMV 46 -LcrV immunization conferred long-term protection. Moreover, immunization with reduced doses of OMV 46 -LcrV exhibited further lower reactogenicity and still provided great protection against pneumonic plague. Our studies strongly demonstrate the feasibility of OMV 46 -LcrV as a new type of plague vaccine candidate.
Collapse
|
40
|
Jackson WD, Giacomassi C, Ward S, Owen A, Luis TC, Spear S, Woollard KJ, Johansson C, Strid J, Botto M. TLR7 activation at epithelial barriers promotes emergency myelopoiesis and lung antiviral immunity. eLife 2023; 12:e85647. [PMID: 37566453 PMCID: PMC10465127 DOI: 10.7554/elife.85647] [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: 12/17/2022] [Accepted: 08/10/2023] [Indexed: 08/12/2023] Open
Abstract
Monocytes are heterogeneous innate effector leukocytes generated in the bone marrow and released into circulation in a CCR2-dependent manner. During infection or inflammation, myelopoiesis is modulated to rapidly meet the demand for more effector cells. Danger signals from peripheral tissues can influence this process. Herein we demonstrate that repetitive TLR7 stimulation via the epithelial barriers drove a potent emergency bone marrow monocyte response in mice. This process was unique to TLR7 activation and occurred independently of the canonical CCR2 and CX3CR1 axes or prototypical cytokines. The monocytes egressing the bone marrow had an immature Ly6C-high profile and differentiated into vascular Ly6C-low monocytes and tissue macrophages in multiple organs. They displayed a blunted cytokine response to further TLR7 stimulation and reduced lung viral load after RSV and influenza virus infection. These data provide insights into the emergency myelopoiesis likely to occur in response to the encounter of single-stranded RNA viruses at barrier sites.
Collapse
Affiliation(s)
- William D Jackson
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
| | - Chiara Giacomassi
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
| | - Sophie Ward
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
| | - Amber Owen
- National Heart and Lung Institute, Imperial College LondonLondonUnited Kingdom
| | - Tiago C Luis
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
| | - Sarah Spear
- Division of Cancer, Department of Surgery and Cancer, Imperial College LondonLondonUnited Kingdom
| | - Kevin J Woollard
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
| | - Cecilia Johansson
- National Heart and Lung Institute, Imperial College LondonLondonUnited Kingdom
| | - Jessica Strid
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
| | - Marina Botto
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
41
|
Huang J, Chen Z, Ye Y, Shao Y, Zhu P, Li X, Ma Y, Xu F, Zhou J, Wu M, Gao X, Yang Y, Zhang J, Hao C. DTX3L Enhances Type I Interferon Antiviral Response by Promoting the Ubiquitination and Phosphorylation of TBK1. J Virol 2023; 97:e0068723. [PMID: 37255478 PMCID: PMC10308958 DOI: 10.1128/jvi.00687-23] [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] [Received: 05/07/2023] [Accepted: 05/11/2023] [Indexed: 06/01/2023] Open
Abstract
Studies already revealed that some E3 ubiquitin ligases participated in the immune response after viral infection by regulating the type I interferon (IFN) pathway. Here, we demonstrated that type I interferon signaling enhanced the translocation of ETS1 to the nucleus and the promoter activity of E3 ubiquitin ligase DTX3L (deltex E3 ubiquitin ligase 3L) after virus infection and thus increased the expression of DTX3L. Further experiments suggested that DTX3L ubiquitinated TBK1 at K30 and K401 sites on K63-linked ubiquitination pathway. DTX3L was also necessary for mediating the phosphorylation of TBK1 through binding with the tyrosine kinase SRC: both together enhanced the activation of TBK1. Therefore, DTX3L, being an important positive-feedback regulator of type I interferon, exerted a key role in antiviral response. IMPORTANCE Our present study evaluated DTX3L as an antiviral molecule by promoting IFN production and establishing an IFN-β-ETS1-DTX3L-TBK1 positive-feedback loop as a novel immunomodulatory step to enhance interferon signaling and inhibit respiratory syncytial virus (RSV) infection. Our finding enriches and complements the biological function of DTX3L and provides a new strategy to protect against lung diseases such as bronchiolitis and pneumonia that develop with RSV.
Collapse
Affiliation(s)
- Jiaqi Huang
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhengrong Chen
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yunfei Ye
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yu Shao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Peijie Zhu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Xiaoping Li
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
- Reproductive Medicine Center, the First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yu Ma
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fei Xu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Ji Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Mengyun Wu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Xiu Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Yi Yang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, People's Republic of China
| | - Chuangli Hao
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
42
|
Britt RD, Ruwanpathirana A, Ford ML, Lewis BW. Macrophages Orchestrate Airway Inflammation, Remodeling, and Resolution in Asthma. Int J Mol Sci 2023; 24:10451. [PMID: 37445635 PMCID: PMC10341920 DOI: 10.3390/ijms241310451] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Asthma is a heterogenous chronic inflammatory lung disease with endotypes that manifest different immune system profiles, severity, and responses to current therapies. Regardless of endotype, asthma features increased immune cell infiltration, inflammatory cytokine release, and airway remodeling. Lung macrophages are also heterogenous in that there are separate subsets and, depending on the environment, different effector functions. Lung macrophages are important in recruitment of immune cells such as eosinophils, neutrophils, and monocytes that enhance allergic inflammation and initiate T helper cell responses. Persistent lung remodeling including mucus hypersecretion, increased airway smooth muscle mass, and airway fibrosis contributes to progressive lung function decline that is insensitive to current asthma treatments. Macrophages secrete inflammatory mediators that induce airway inflammation and remodeling. Additionally, lung macrophages are instrumental in protecting against pathogens and play a critical role in resolution of inflammation and return to homeostasis. This review summarizes current literature detailing the roles and existing knowledge gaps for macrophages as key inflammatory orchestrators in asthma pathogenesis. We also raise the idea that modulating inflammatory responses in lung macrophages is important for alleviating asthma.
Collapse
Affiliation(s)
- Rodney D Britt
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Anushka Ruwanpathirana
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43205, USA
| | - Maria L Ford
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43205, USA
| | - Brandon W Lewis
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
| |
Collapse
|
43
|
Ghimire R, Shrestha R, Amaradhi R, Patton T, Whitley C, Chanda D, Liu L, Ganesh T, More S, Channappanavar R. Toll-like receptor 7 (TLR7)-mediated antiviral response protects mice from lethal SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539929. [PMID: 37214943 PMCID: PMC10197544 DOI: 10.1101/2023.05.08.539929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
SARS-CoV-2-induced impaired antiviral and excessive inflammatory responses cause fatal pneumonia. However, the key pattern recognition receptors that elicit effective antiviral and lethal inflammatory responses in-vivo are not well defined. CoVs possess single-stranded RNA (ssRNA) genome that is abundantly produced during infection and stimulates both antiviral interferon (IFN) and inflammatory cytokine/ chemokine responses. Therefore, in this study, using wild-type control and TLR7 deficient BALB/c mice infected with a mouse-adapted SARS-COV-2 (MA-CoV-2), we evaluated the role of TLR7 signaling in MA-CoV-2-induced antiviral and inflammatory responses and disease outcome. We show that TLR7-deficient mice are more susceptible to MA-CoV-2 infection as compared to infected control mice. Further evaluation of MA-CoV-2 infected lungs showed significantly reduced mRNA levels of antiviral type I (IFNα/β) and type III (IFNλ) IFNs, IFN stimulated genes (ISGs, ISG15 and CXCL10), and several pro-inflammatory cytokines/chemokines in TLR7 deficient compared to control mice. Reduced lung IFN/ISG levels and increased morbidity/mortality in TLR7 deficient mice correlated with high lung viral titer. Detailed examination of total cells from MA-CoV-2 infected lungs showed high neutrophil count in TLR7 deficient mice compared to control mice. Additionally, blocking TLR7 activity post-MA-CoV-2 infection using a specific inhibitor also enhanced disease severity. In summary, our results conclusively establish that TLR7 signaling is protective during SARS-CoV-2 infection, and despite robust inflammatory response, TLR7-mediated IFN/ISG responses likely protect the host from lethal disease. Given similar outcomes in control and TLR7 deficient humans and mice, these results show that MA-CoV-2 infected mice serve as excellent model to study COVID-19.
Collapse
|
44
|
T'Jonck W, Bain CC. The role of monocyte-derived macrophages in the lung: it's all about context. Int J Biochem Cell Biol 2023; 159:106421. [PMID: 37127181 DOI: 10.1016/j.biocel.2023.106421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
Macrophages are present in every tissue of the body where they play crucial roles in maintaining tissue homeostasis and providing front line defence against pathogens. Arguably, this is most important at mucosal barrier tissues, such as the lung and gut, which are major ports of entry for pathogens. However, a common feature of inflammation, infection or injury is the loss of tissue resident macrophages and accumulation of monocytes from the circulation, which differentiate, to different extents, into macrophages. The exact fate and function of these elicited, monocyte-derived macrophages in infection, injury and inflammation remains contentious. While some studies have documented the indispensable nature of monocytes and their macrophage derivatives in combatting infection and restoration of lung homeostasis following insult, observations from clinical studies and preclinical models of lung infection/injury shows that monocytes and their progeny can become dysregulated in severe pathology, often perpetuating rather than resolving the insult. In this Mini Review, we aim to bring together these somewhat contradictory reports by discussing how the plasticity of monocytes allow them to assume distinct functions in different contexts in the lung, from health to infection, and effective tissue repair to fibrotic disease.
Collapse
Affiliation(s)
- Wouter T'Jonck
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, EH16 4TJ, U.K; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter
| | - Calum C Bain
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, EH16 4TJ, U.K; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh BioQuarter
| |
Collapse
|
45
|
Labzin LI, Chew KY, Eschke K, Wang X, Esposito T, Stocks CJ, Rae J, Patrick R, Mostafavi H, Hill B, Yordanov TE, Holley CL, Emming S, Fritzlar S, Mordant FL, Steinfort DP, Subbarao K, Nefzger CM, Lagendijk AK, Gordon EJ, Parton RG, Short KR, Londrigan SL, Schroder K. Macrophage ACE2 is necessary for SARS-CoV-2 replication and subsequent cytokine responses that restrict continued virion release. Sci Signal 2023; 16:eabq1366. [PMID: 37098119 DOI: 10.1126/scisignal.abq1366] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Macrophages are key cellular contributors to the pathogenesis of COVID-19, the disease caused by the virus SARS-CoV-2. The SARS-CoV-2 entry receptor ACE2 is present only on a subset of macrophages at sites of SARS-CoV-2 infection in humans. Here, we investigated whether SARS-CoV-2 can enter macrophages, replicate, and release new viral progeny; whether macrophages need to sense a replicating virus to drive cytokine release; and, if so, whether ACE2 is involved in these mechanisms. We found that SARS-CoV-2 could enter, but did not replicate within, ACE2-deficient human primary macrophages and did not induce proinflammatory cytokine expression. By contrast, ACE2 overexpression in human THP-1-derived macrophages permitted SARS-CoV-2 entry, processing and replication, and virion release. ACE2-overexpressing THP-1 macrophages sensed active viral replication and triggered proinflammatory, antiviral programs mediated by the kinase TBK-1 that limited prolonged viral replication and release. These findings help elucidate the role of ACE2 and its absence in macrophage responses to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Larisa I Labzin
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - Kathrin Eschke
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiaohui Wang
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, QLD 4072, Australia
| | - Tyron Esposito
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, QLD 4072, Australia
| | - Claudia J Stocks
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, QLD 4072, Australia
| | - James Rae
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD 4072, Australia
| | - Ralph Patrick
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Helen Mostafavi
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Brittany Hill
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Teodor E Yordanov
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Caroline L Holley
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, QLD 4072, Australia
| | - Stefan Emming
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, QLD 4072, Australia
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Francesca L Mordant
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Daniel P Steinfort
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Christian M Nefzger
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Anne K Lagendijk
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Emma J Gordon
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD 4072, Australia
| | - Kirsty R Short
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), University of Queensland, Brisbane, QLD 4072, Australia
- IMB Centre for Inflammation and Disease Research, University of Queensland, Brisbane, QLD 4072, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
46
|
Feng C, Jin C, Liu K, Yang Z. Microbiota-derived short chain fatty acids: Their role and mechanisms in viral infections. Biomed Pharmacother 2023. [DOI: 10.1016/j.biopha.2023.114414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023] Open
|
47
|
Wang L, Wang D, Zhang T, Ma Y, Tong X, Fan H. The role of immunometabolism in macrophage polarization and its impact on acute lung injury/acute respiratory distress syndrome. Front Immunol 2023; 14:1117548. [PMID: 37020557 PMCID: PMC10067752 DOI: 10.3389/fimmu.2023.1117548] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/03/2023] [Indexed: 04/07/2023] Open
Abstract
Lung macrophages constitute the first line of defense against airborne particles and microbes and are key to maintaining pulmonary immune homeostasis. There is increasing evidence suggesting that macrophages also participate in the pathogenesis of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), including the modulation of inflammatory responses and the repair of damaged lung tissues. The diversity of their functions may be attributed to their polarized states. Classically activated or inflammatory (M1) macrophages and alternatively activated or anti-inflammatory (M2) macrophages are the two main polarized macrophage phenotypes. The precise regulatory mechanism of macrophage polarization is a complex process that is not completely understood. A growing body of literature on immunometabolism has demonstrated the essential role of immunometabolism and its metabolic intermediates in macrophage polarization. In this review, we summarize macrophage polarization phenotypes, the role of immunometabolism, and its metabolic intermediates in macrophage polarization and ALI/ARDS, which may represent a new target and therapeutic direction.
Collapse
Affiliation(s)
- Lian Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Dongguang Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Tianli Zhang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Ma
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Abstract
Mucosal tissues are constantly exposed to the outside environment. They receive signals from the commensal microbiome and tissue-specific triggers including alimentary and airborne elements and are tasked to maintain balance in the absence of inflammation and infection. Here, we present neutrophils as sentinel cells in mucosal immunity. We discuss the roles of neutrophils in mucosal homeostasis and overview clinical susceptibilities in patients with neutrophil defects. Finally, we present concepts related to specification of neutrophil responses within specific mucosal tissue microenvironments.
Collapse
Affiliation(s)
- Lakmali M. Silva
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Tae Sung Kim
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Niki M. Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
49
|
Inflammatory cell death: how macrophages sense neighbouring cell infection and damage. Biochem Soc Trans 2023; 51:303-313. [PMID: 36695550 PMCID: PMC9987993 DOI: 10.1042/bst20220807] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Programmed cell death is a critical host defence strategy during viral infection. Neighbouring cells deal with this death in distinct ways depending on how the infected cell dies. While apoptosis is considered immunologically silent, the lytic pathways of necroptosis and pyroptosis trigger inflammatory responses by releasing inflammatory host molecules. All these pathways have been implicated in influenza A virus infection. Here, we review how cells sense neighbouring infection and death and how sensing shapes ensuing inflammatory responses.
Collapse
|
50
|
Korkmaz FT, Traber KE. Innate immune responses in pneumonia. Pneumonia (Nathan) 2023; 15:4. [PMID: 36829255 PMCID: PMC9957695 DOI: 10.1186/s41479-023-00106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/05/2023] [Indexed: 02/26/2023] Open
Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology & Infectious Disease, University of Massachusetts, Worcester, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|