1
|
Eddens T, Parks OB, Zhang Y, Manni ML, Casanova JL, Ogishi M, Williams JV. PD-1 signaling in neonates restrains CD8 + T cell function and protects against respiratory viral immunopathology. Mucosal Immunol 2024; 17:476-490. [PMID: 38176655 PMCID: PMC11180597 DOI: 10.1016/j.mucimm.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Respiratory viral infections, including human metapneumovirus (HMPV), remain a leading cause of morbidity and mortality in neonates and infants. However, the mechanisms behind the increased sensitivity to those respiratory viral infections in neonates are poorly understood. Neonates, unlike adults, have several anti-inflammatory mechanisms in the lung, including elevated baseline expression of programmed death ligand 1 (PD-L1), a ligand for the inhibitory receptor programmed cell death protein 1 (PD-1). We thus hypothesized that neonates would rely on PD-1:PD-L1 signaling to restrain antiviral CD8 responses. To test this, we developed a neonatal primary HMPV infection model using wild-type C57BL/6 (B6) and Pdcd1-/- (lacking PD-1) mice. HMPV-infected neonatal mice had increased PD-L1/PD-L2 co-expression on innate immune cells but a similar number of antigen-specific CD8+ T cells and upregulation of PD-1 to that of adult B6 mice. Neonatal CD8+ T cells had reduced interferon-gamma (IFN-γ), granzyme B, and interleukin-2 production compared with B6 adults. Pdcd1-/- neonatal CD8+ T cells had markedly increased production of IFN-γ and granzyme B compared with B6 neonates. Pdcd1-/- neonates had increased acute pathology with HMPV or influenza. Pdcd1-/- neonates infected with HMPV had long-term changes in pulmonary physiology with evidence of immunopathology and a persistent CD8+ T-cell response with increased granzyme B production. Using single-cell ribonucleic acid sequencing from a child lacking PD-1 signaling, a similar activated CD8+ T-cell signature with increased granzyme B expression was observed. These data indicate that PD-1 signaling critically limits CD8+ T-cell effector functions and prevents immunopathology in response to neonatal respiratory viral infections.
Collapse
Affiliation(s)
- Taylor Eddens
- Division of Allergy and Immunology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA; University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, Pennsylvania, USA
| | - Olivia B Parks
- University of Pittsburgh Medical Scientist Training Program, Pittsburgh, Pennsylvania, USA
| | - Yu Zhang
- Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, Pennsylvania, USA
| | - Michelle L Manni
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France; University Paris Cité, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA; Howard Hughes Medical Institute, New York, New York, USA; Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, New York, USA
| | - John V Williams
- University of Pittsburgh School of Medicine, Department of Pediatrics, Pittsburgh, Pennsylvania, USA; Institute for Infection, Inflammation, and Immunity in Children (i4Kids), Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
2
|
DaMata JP, Zelkoski AE, Nhan PB, Ennis KHE, Kim JS, Lu Z, Malloy AMW. Dissociation protocols influence the phenotypes of lymphocyte and myeloid cell populations isolated from the neonatal lymph node. Front Immunol 2024; 15:1368118. [PMID: 38756770 PMCID: PMC11097666 DOI: 10.3389/fimmu.2024.1368118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Frequencies and phenotypes of immune cells differ between neonates and adults in association with age-specific immune responses. Lymph nodes (LN) are critical tissue sites to quantify and define these differences. Advances in flow cytometry have enabled more multifaceted measurements of complex immune responses. Tissue processing can affect the immune cells under investigation that influence key findings. To understand the impact on immune cells in the LN after processing for single-cell suspension, we compared three dissociation protocols: enzymatic digestion, mechanical dissociation with DNase I treatment, and mechanical dissociation with density gradient separation. We analyzed cell yields, viability, phenotypic and maturation markers of immune cells from the lung-draining LN of neonatal and adult mice two days after intranasal respiratory syncytial virus (RSV) infection. While viability was consistent across age groups, the protocols influenced the yield of subsets defined by important phenotypic and activation markers. Moreover, enzymatic digestion did not show higher overall yields of conventional dendritic cells and macrophages from the LN. Together, our findings show that the three dissociation protocols have similar impacts on the number and viability of cells isolated from the neonatal and adult LN. However, enzymatic digestion impacts the mean fluorescence intensity of key lineage and activation markers that may influence experimental findings.
Collapse
Affiliation(s)
- Jarina P. DaMata
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Amanda E. Zelkoski
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| | - Paula B. Nhan
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Katherine H. E. Ennis
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Ji Sung Kim
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Zhongyan Lu
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Allison M. W. Malloy
- Laboratory of Infectious Diseases and Host Defense, Department of Pediatrics, Uniformed Services University of Health Sciences (USUHS), Bethesda, MD, United States
| |
Collapse
|
3
|
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
|
4
|
Smits HH, Jochems SP. Diverging patterns in innate immunity against respiratory viruses during a lifetime: lessons from the young and the old. Eur Respir Rev 2024; 33:230266. [PMID: 39009407 PMCID: PMC11262623 DOI: 10.1183/16000617.0266-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/16/2024] [Indexed: 07/17/2024] Open
Abstract
Respiratory viral infections frequently lead to severe respiratory disease, particularly in vulnerable populations such as young children, individuals with chronic lung conditions and older adults, resulting in hospitalisation and, in some cases, fatalities. The innate immune system plays a crucial role in monitoring for, and initiating responses to, viruses, maintaining a state of preparedness through the constant expression of antimicrobial defence molecules. Throughout the course of infection, innate immunity remains actively involved, contributing to viral clearance and damage control, with pivotal contributions from airway epithelial cells and resident and newly recruited immune cells. In instances where viral infections persist or are not effectively eliminated, innate immune components prominently contribute to the resulting pathophysiological consequences. Even though both young children and older adults are susceptible to severe respiratory disease caused by various respiratory viruses, the underlying mechanisms may differ significantly. Children face the challenge of developing and maturing their immunity, while older adults contend with issues such as immune senescence and inflammaging. This review aims to compare the innate immune responses in respiratory viral infections across both age groups, identifying common central hubs that could serve as promising targets for innovative therapeutic and preventive strategies, despite the apparent differences in underlying mechanisms.
Collapse
Affiliation(s)
- Hermelijn H Smits
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P Jochems
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Dieterlen MT, Messer EK, Klaeske K, Sieg F, Eifert S, Haunschild J, Jawad K, Saeed D, Dashkevich A, Borger MA. Pre-Implant Immune Status is Associated with Infection Risk After Left Ventricular Assist Device Implantation. J Inflamm Res 2024; 17:581-589. [PMID: 38318245 PMCID: PMC10840410 DOI: 10.2147/jir.s424879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 11/10/2023] [Indexed: 02/07/2024] Open
Abstract
Purpose Infection is the most common complication after left ventricular assist device (LVAD) implantation. The immune status of LVAD patients is relevant for the incidence and severity of infection, but it is unknown if there is a predisposing immune status prior to LVAD implantation that contributes to an increased risk for infection in the post-implant period. We analyzed the pre-LVAD immune status in patients with infection within 3 months after LVAD implantation in comparison to infection-free patients. Patients and Methods Fifty-four consecutive LVAD patients were included in this study. According to their infectious history in the first 3 months after LVAD implantation, these patients were grouped into an infection (n=23) and an infection-free group (n=31). Pre-LVAD blood samples were obtained for flow cytometric analysis of immunological parameters including B cells, subsets of T, dendritic and natural killer cells. Patient-specific, clinical and laboratory data were recorded. Results Blood count analysis prior to LVAD implantation showed comparable counts of erythrocytes (p=0.19), platelets (p=0.33) and leukocytes (p=0.50) between patients with infection and infection-free patients in the post-implant period. Patients with infection in the first 3 months after LVAD implantation had lower concentrations of lymphocytes (p=0.02). Forty percent of the patients with infection showed more often pre-LVAD neutrophil-to-lymphocyte ratios (NLR) >7 than patients without infection in the first 3 months after LVAD implantation (14%, p=0.05). Patients with infection already had lower percentages of CD3+ T cells (p=0.03), CD19+ B cells (p<0.01), BDCA2+ pDCs (p=0.03) and BDCA4+ plasmacytoid DCs (pDCs) (p=0.05) prior to LVAD implantation than infection-free patients. Conclusion Our results demonstrated that patients with infection in the early post-implant period showed lower concentrations of lymphocytes, especially of CD3+ T cells and CD19+ B cells, decreased percentages of BDCA2+ and BDCA4+ pDCs, and had more often NLRs >7 indicating moderate-to-severe inflammation. Thus, we identified specific immunological changes pre-LVAD that could help to identify patients at risk for infection in the early post-implant period.
Collapse
Affiliation(s)
- Maja-Theresa Dieterlen
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Eva Katharina Messer
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Kristin Klaeske
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Franz Sieg
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Sandra Eifert
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Josephina Haunschild
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Khalil Jawad
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Diyar Saeed
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Alexey Dashkevich
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| | - Michael Andrew Borger
- University Clinic of Cardiac Surgery, Leipzig Heart Center, HELIOS Clinic, Leipzig, Germany
| |
Collapse
|
6
|
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
|
7
|
Dagenais A, Villalba-Guerrero C, Olivier M. Trained immunity: A “new” weapon in the fight against infectious diseases. Front Immunol 2023; 14:1147476. [PMID: 36993966 PMCID: PMC10040606 DOI: 10.3389/fimmu.2023.1147476] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
Innate immune cells can potentiate the response to reinfection through an innate form of immunological memory known as trained immunity. The potential of this fast-acting, nonspecific memory compared to traditional adaptive immunological memory in prophylaxis and therapy has been a topic of great interest in many fields, including infectious diseases. Amidst the rise of antimicrobial resistance and climate change—two major threats to global health—, harnessing the advantages of trained immunity compared to traditional forms of prophylaxis and therapy could be game-changing. Here, we present recent works bridging trained immunity and infectious disease that raise important discoveries, questions, concerns, and novel avenues for the modulation of trained immunity in practice. By exploring the progress in bacterial, viral, fungal, and parasitic diseases, we equally highlight future directions with a focus on particularly problematic and/or understudied pathogens.
Collapse
Affiliation(s)
- Amy Dagenais
- Department of Microbiology and Immunology, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Carlos Villalba-Guerrero
- Department of Microbiology and Immunology, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
- *Correspondence: Martin Olivier,
| |
Collapse
|
8
|
Tovo PA, Garazzino S, Savino F, Daprà V, Pruccoli G, Dini M, Filisetti G, Funiciello E, Galliano I, Bergallo M. Expressions of Type I and III Interferons, Endogenous Retroviruses, TRIM28, and SETDB1 in Children with Respiratory Syncytial Virus Bronchiolitis. Curr Issues Mol Biol 2023; 45:1197-1217. [PMID: 36826024 PMCID: PMC9954910 DOI: 10.3390/cimb45020079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Interferons (IFNs) and IFN-stimulated genes (ISGs) play essential roles for the control of viral infections. Their expression in infants with respiratory syncytial virus (RSV) bronchiolitis is poorly defined. Human endogenous retroviruses (HERVs) represent 8% of our genome and modulate inflammatory and immune reactions. TRIM28 and SETDB1 participate in the epigenetic regulation of genes involved in the immune response, including IFNs and HERVs. No study has explored the expression of HERVs, TRIM28, and SETDB1 during RSV bronchiolitis. We assessed, through a PCR real-time Taqman amplification assay, the transcription levels of six IFN-I ISGs, four IFNλs, the pol genes of HERV-H, -K, and -W families, the env genes of Syncytin (SYN)1 and SYN2, and of TRIM28/SETDB1 in whole blood from 37 children hospitalized for severe RSV bronchiolitis and in healthy children (HC). The expression of most IFN-I ISGs was significantly higher in RSV+ patients than in age-matched HC, but it was inhibited by steroid therapy. The mRNA concentrations of IFN-λs were comparable between patients and age-matched HC. This lack of RSV-driven IFN-III activation may result in the defective protection of the airway mucosal surface leading to severe bronchiolitis. The expression of IFN-III showed a positive correlation with age in HC, that could account for the high susceptibility of young children to viral respiratory tract infections. The transcription levels of every HERV gene were significantly lower in RSV+ patients than in HC, while the expressions of TRIM28/SETDB1 were overlapping. Given the negative impact of HERVs and the positive effects of TRIM28/SETDB1 on innate and adaptive immune responses, the downregulation of the former and the normal expression of the latter may contribute to preserving immune functions against infection.
Collapse
Affiliation(s)
- Pier-Angelo Tovo
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Correspondence: Correspondence: (P.-A.T.); (M.B.)
| | - Silvia Garazzino
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Francesco Savino
- Early Infancy Special Care Unit, Department of Pediatric Care, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Valentina Daprà
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Giulia Pruccoli
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Maddalena Dini
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Giacomo Filisetti
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Early Infancy Special Care Unit, Department of Pediatric Care, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Elisa Funiciello
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Infectious Diseases Unit, Department of Pediatrics, Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy
| | - Ilaria Galliano
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
| | - Massimiliano Bergallo
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Pediatric Laboratory, Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, 10126 Turin, Italy
- Correspondence: Correspondence: (P.-A.T.); (M.B.)
| |
Collapse
|
9
|
Bergeron HC, Kauvar LM, Tripp RA. Anti-G protein antibodies targeting the RSV G protein CX3C chemokine region improve the interferon response. Ther Adv Infect Dis 2023; 10:20499361231161157. [PMID: 36938145 PMCID: PMC10017941 DOI: 10.1177/20499361231161157] [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: 09/07/2022] [Accepted: 02/01/2023] [Indexed: 03/15/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is a poor inducer of antiviral interferon (IFN) responses which result in incomplete immunity and RSV disease. Several RSV proteins alter antiviral responses, including the non-structural proteins (NS1, NS2) and the major viral surface proteins, that is, fusion (F) and attachment (G) proteins. The G protein modifies the host immune response to infection linked in part through a CX3 C chemokine motif. Anti-G protein monoclonal antibodies (mAbs), that is, clones 3D3 and 2D10 that target the G protein CX3C chemokine motif can neutralize RSV and inhibit G protein-CX3CR1 mediated chemotaxis. Objectives Determine how monoclonal antibodies against the RSV F and G proteins modify the type I and III IFN responses to RSV infection. Design As the G protein CX3 C motif is implicated in IFN antagonism, we evaluated two mAbs that block G protein CX3C-CX3CR1 interaction and compared responses to isotype mAb control using a functional cellular assay and mouse model. Methods Mouse lung epithelial cells (MLE-15 cells) and BALB/c mice were infected with RSV Line19 F following prophylactic mAb treatment. Cell supernatant or bronchoalveolar lavage fluid (BALF) were assayed for types I and III IFNs. Cells were interrogated for changes in IFN-related gene expression. Results Treatment with an anti-G protein mAb (3D3) resulted in improved IFN responses compared with isotype control following infection with RSV, partially independently of neutralization, and this was linked to upregulated SOCS1 expression. Conclusions These findings show that anti-G protein antibodies improve the protective early antiviral response, which has important implications for vaccine and therapeutic design. Plain Language Summary RSV is a leading cause of respiratory disease in infants and the elderly. The only Food and Drug Administration-approved prophylactic treatment is limited to an anti-F protein monoclonal antibody (mAb), that is, palivizumab which has modest efficacy against RSV disease. Accumulating evidence suggests that targeting the RSV attachment (G) protein may provide improved protection from RSV disease. It is known that the G protein is an IFN antagonist, and IFN has been shown to be protective against RSV disease. In this study, we compared IFN responses in mouse lung epithelial (MLE-15) cells and in mice infected with RSV Line19 F treated with anti-G protein or anti-F protein mAbs. The levels of type I and III IFNs were determined. Anti-G protein mAbs improved the levels of IFNs compared with isotype-treated controls. These findings support the concept that anti-G protein mAbs mediate improved IFN responses against RSV disease, which may enable improved treatment of RSV infections.
Collapse
Affiliation(s)
- Harrison C. Bergeron
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | | | |
Collapse
|
10
|
Pieren DKJ, Boer MC, de Wit J. The adaptive immune system in early life: The shift makes it count. Front Immunol 2022; 13:1031924. [PMID: 36466865 PMCID: PMC9712958 DOI: 10.3389/fimmu.2022.1031924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/31/2022] [Indexed: 10/13/2023] Open
Abstract
Respiratory infectious diseases encountered early in life may result in life-threatening disease in neonates, which is primarily explained by the relatively naive neonatal immune system. Whereas vaccines are not readily available for all infectious diseases, vaccinations have greatly reduced childhood mortality. However, repeated vaccinations are required to reach protective immunity in infants and not all vaccinations are effective at young age. Moreover, protective adaptive immunity elicited by vaccination wanes more rapidly at young age compared to adulthood. The infant adaptive immune system has previously been considered immature but this paradigm has changed during the past years. Recent evidence shows that the early life adaptive immune system is equipped with a strong innate-like effector function to eliminate acute pathogenic threats. These strong innate-like effector capacities are in turn kept in check by a tolerogenic counterpart of the adaptive system that may have evolved to maintain balance and to reduce collateral damage. In this review, we provide insight into these aspects of the early life's adaptive immune system by addressing recent literature. Moreover, we speculate that this shift from innate-like and tolerogenic adaptive immune features towards formation of immune memory may underlie different efficacy of infant vaccination in these different phases of immune development. Therefore, presence of innate-like and tolerogenic features of the adaptive immune system may be used as a biomarker to improve vaccination strategies against respiratory and other infections in early life.
Collapse
Affiliation(s)
| | | | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
11
|
Kumova OK, Galani IE, Rao A, Johnson H, Triantafyllia V, Matt SM, Pascasio J, Gaskill PJ, Andreakos E, Katsikis PD, Carey AJ. Severity of neonatal influenza infection is driven by type I interferon and oxidative stress. Mucosal Immunol 2022; 15:1309-1320. [PMID: 36352099 PMCID: PMC9724789 DOI: 10.1038/s41385-022-00576-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 09/26/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022]
Abstract
Neonates exhibit increased susceptibility to respiratory viral infections, attributed to inflammation at the developing pulmonary air-blood interface. IFN I are antiviral cytokines critical to control viral replication, but also promote inflammation. Previously, we established a neonatal murine influenza virus (IV) model, which demonstrates increased mortality. Here, we sought to determine the role of IFN I in this increased mortality. We found that three-day-old IFNAR-deficient mice are highly protected from IV-induced mortality. In addition, exposure to IFNβ 24 h post IV infection accelerated death in WT neonatal animals but did not impact adult mortality. In contrast, IFN IIIs are protective to neonatal mice. IFNβ induced an oxidative stress imbalance specifically in primary neonatal IV-infected pulmonary type II epithelial cells (TIIEC), not in adult TIIECs. Moreover, neonates did not have an infection-induced increase in antioxidants, including a key antioxidant, superoxide dismutase 3, as compared to adults. Importantly, antioxidant treatment rescued IV-infected neonatal mice, but had no impact on adult morbidity. We propose that IFN I exacerbate an oxidative stress imbalance in the neonate because of IFN I-induced pulmonary TIIEC ROS production coupled with developmentally regulated, defective antioxidant production in response to IV infection. This age-specific imbalance contributes to mortality after respiratory infections in this vulnerable population.
Collapse
Affiliation(s)
- Ogan K. Kumova
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Ioanna-Evdokia Galani
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Abhishek Rao
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Hannah Johnson
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Vasiliki Triantafyllia
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Stephanie M. Matt
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Judy Pascasio
- Pathology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Peter J. Gaskill
- Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Peter D. Katsikis
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alison J. Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States.,Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
12
|
Eddens T, Parks OB, Williams JV. Neonatal Immune Responses to Respiratory Viruses. Front Immunol 2022; 13:863149. [PMID: 35493465 PMCID: PMC9047724 DOI: 10.3389/fimmu.2022.863149] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Respiratory tract infections are a leading cause of morbidity and mortality in newborns, infants, and young children. These early life infections present a formidable immunologic challenge with a number of possibly conflicting goals: simultaneously eliminate the acute pathogen, preserve the primary gas-exchange function of the lung parenchyma in a developing lung, and limit long-term sequelae of both the infection and the inflammatory response. The latter has been most well studied in the context of childhood asthma, where multiple epidemiologic studies have linked early life viral infection with subsequent bronchospasm. This review will focus on the clinical relevance of respiratory syncytial virus (RSV), human metapneumovirus (HMPV), and rhinovirus (RV) and examine the protective and pathogenic host responses within the neonate.
Collapse
Affiliation(s)
- Taylor Eddens
- Pediatric Scientist Development Program, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Division of Allergy/Immunology, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Olivia B. Parks
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - John V. Williams
- Division of Pediatric Infectious Diseases, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
13
|
Feng Z, Xu L, Xie Z. Receptors for Respiratory Syncytial Virus Infection and Host Factors Regulating the Life Cycle of Respiratory Syncytial Virus. Front Cell Infect Microbiol 2022; 12:858629. [PMID: 35281439 PMCID: PMC8913501 DOI: 10.3389/fcimb.2022.858629] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 12/02/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of lower respiratory tract infections and responsible for a large proportion of mortality in children and the elderly. There are no licensed vaccines available to date. Prophylaxis and therapeutic RSV-specific antibodies are limited to populations at high risk owing to high cost and uncertain clinical value. Receptors and host factors are two determinants important for virus entry and establishment of infection in vivo. The identification and understanding of viral receptors and host factors can help us to gain insight into the pathogenesis of RSV infection. Herein, we reviewed receptors and host factors that have been reported thus far. RSV could bind to CX3C chemokine receptor 1 and heparan sulfate proteoglycans via the G protein, and to nucleolin, insulin-like growth factor-1 receptor, epidermal growth factor, and intercellular adhesion molecule-1 via the F protein. Seven host restriction factors and 13 host factors essential for RSV infection were reviewed. We characterized the functions and their roles in the life cycle of RSV, trying to provide an update on the information of RSV-related receptors and host factors.
Collapse
Affiliation(s)
- 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), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, 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), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Lili Xu,
| | - 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), Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
14
|
La muqueuse pulmonaire en période périnatale : un monde à comprendre pour lutter contre la sensibilité du jeune à la bronchiolite. Rev Mal Respir 2022; 39:104-107. [DOI: 10.1016/j.rmr.2022.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/19/2022]
|
15
|
Descamps D, Peres de Oliveira A, Gonnin L, Madrières S, Fix J, Drajac C, Marquant Q, Bouguyon E, Pietralunga V, Iha H, Morais Ventura A, Tangy F, Vidalain PO, Eléouët JF, Galloux M. Depletion of TAX1BP1 Amplifies Innate Immune Responses during Respiratory Syncytial Virus Infection. J Virol 2021; 95:e0091221. [PMID: 34431698 PMCID: PMC8549506 DOI: 10.1128/jvi.00912-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the main cause of acute respiratory infections in young children and also has a major impact on the elderly and immunocompromised people. In the absence of a vaccine or efficient treatment, a better understanding of RSV interactions with the host antiviral response during infection is needed. Previous studies revealed that cytoplasmic inclusion bodies (IBs), where viral replication and transcription occur, could play a major role in the control of innate immunity during infection by recruiting cellular proteins involved in the host antiviral response. We recently showed that the morphogenesis of IBs relies on a liquid-liquid-phase separation mechanism depending on the interaction between viral nucleoprotein (N) and phosphoprotein (P). These scaffold proteins are expected to play a central role in the recruitment of cellular proteins to IBs. Here, we performed a yeast two-hybrid screen using RSV N protein as bait and identified the cellular protein TAX1BP1 as a potential partner of this viral protein. This interaction was validated by pulldown and immunoprecipitation assays. We showed that TAX1BP1 suppression has only a limited impact on RSV infection in cell cultures. However, RSV replication is decreased in TAX1BP1-deficient (TAX1BP1 knockout [TAX1BP1KO]) mice, whereas the production of inflammatory and antiviral cytokines is enhanced. In vitro infection of wild-type or TAX1BP1KO alveolar macrophages confirmed that the innate immune response to RSV infection is enhanced in the absence of TAX1BP1. Altogether, our results suggest that RSV could hijack TAX1BP1 to restrain the host immune response during infection. IMPORTANCE Respiratory syncytial virus (RSV), which is the leading cause of lower respiratory tract illness in infants, remains a medical problem in the absence of a vaccine or efficient treatment. This virus is also recognized as a main pathogen in the elderly and immunocompromised people, and the occurrence of coinfections (with other respiratory viruses and bacteria) amplifies the risks of developing respiratory distress. In this context, a better understanding of the pathogenesis associated with viral respiratory infections, which depends on both viral replication and the host immune response, is needed. The present study reveals that the cellular protein TAX1BP1, which interacts with the RSV nucleoprotein N, participates in the control of the innate immune response during RSV infection, suggesting that the N-TAX1BP1 interaction represents a new target for the development of antivirals.
Collapse
Affiliation(s)
| | - Andressa Peres de Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Lorène Gonnin
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Sarah Madrières
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Jenna Fix
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Carole Drajac
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Quentin Marquant
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Edwige Bouguyon
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | | | - Hidekatsu Iha
- Department of Infectious Diseases, Faculty of Medicine, Oita University Idaiga-oka, Hasama Yufu, Japan
| | - Armando Morais Ventura
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Frédéric Tangy
- Unité de Génomique Virale et Vaccination, Institut Pasteur, CNRS UMR-3569, Paris, France
| | - Pierre-Olivier Vidalain
- Unité de Génomique Virale et Vaccination, Institut Pasteur, CNRS UMR-3569, Paris, France
- CIRI, Centre International de Recherche en Infectiologie, Université Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | | | - Marie Galloux
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| |
Collapse
|
16
|
Pruner KB, Pepper M. Local memory CD4 T cell niches in respiratory viral infection. J Exp Med 2021; 218:212432. [PMID: 34160551 PMCID: PMC8225681 DOI: 10.1084/jem.20201733] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/24/2021] [Accepted: 06/08/2021] [Indexed: 12/26/2022] Open
Abstract
Respiratory viral infections present a major threat to global health and prosperity. Over the past century, several have developed into crippling pandemics, including the SARS-CoV-2 virus. Although the generation of neutralizing serum antibodies in response to natural immunity and vaccination are considered to be hallmarks of viral immune protection, antibodies from long-lived plasma cells are subject to immune escape from heterologous clades of zoonotic, recombined, or mutated viruses. Local immunity in the lung can be generated through resident memory immune subsets that rapidly respond to secondary infection and protect from heterologous infection. Although many immune cells are required to achieve the phenomenon of resident memory, herein we highlight the pleiotropic functions of CD4 tissue resident memory T cells in the lung and discuss the implications of resident memory for vaccine design.
Collapse
Affiliation(s)
- Kurt B Pruner
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
17
|
Feng E, Balint E, Poznanski SM, Ashkar AA, Loeb M. Aging and Interferons: Impacts on Inflammation and Viral Disease Outcomes. Cells 2021; 10:708. [PMID: 33806810 PMCID: PMC8004738 DOI: 10.3390/cells10030708] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/16/2022] Open
Abstract
As highlighted by the COVID-19 global pandemic, elderly individuals comprise the majority of cases of severe viral infection outcomes and death. A combined inability to control viral replication and exacerbated inflammatory immune activation in elderly patients causes irreparable immune-mediated tissue pathology in response to infection. Key to these responses are type I, II, and III interferons (IFNs), which are involved in inducing an antiviral response, as well as controlling and suppressing inflammation and immunopathology. IFNs support monocyte/macrophage-stimulated immune responses that clear infection and promote their immunosuppressive functions that prevent excess inflammation and immune-mediated pathology. The timing and magnitude of IFN responses to infection are critical towards their immunoregulatory functions and ability to prevent immunopathology. Aging is associated with multiple defects in the ability of macrophages and dendritic cells to produce IFNs in response to viral infection, leading to a dysregulation of inflammatory immune responses. Understanding the implications of aging on IFN-regulated inflammation will give critical insights on how to treat and prevent severe infection in vulnerable individuals. In this review, we describe the causes of impaired IFN production in aging, and the evidence to suggest that these impairments impact the regulation of the innate and adaptive immune response to infection, thereby causing disease pathology.
Collapse
Affiliation(s)
| | | | | | - Ali A. Ashkar
- Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada; (E.F.); (E.B.); (S.M.P.); (M.L.)
| | | |
Collapse
|
18
|
Shilovskiy IP, Yumashev KV, Nikolsky AA, Vishnyakova LI, Khaitov MR. Molecular and Cellular Mechanisms of Respiratory Syncytial Viral Infection: Using Murine Models to Understand Human Pathology. BIOCHEMISTRY. BIOKHIMIIA 2021; 86:290-306. [PMID: 33838630 PMCID: PMC7957450 DOI: 10.1134/s0006297921030068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/18/2020] [Accepted: 10/18/2020] [Indexed: 12/28/2022]
Abstract
Respiratory syncytial virus (RSV) causes severe pathology of the lower respiratory tract in infants, immunocompromised people, and elderly. Despite decades of research, there is no licensed vaccine against RSV, and many therapeutic drugs are still under development. Detailed understanding of molecular and cellular mechanisms of the RSV infection pathology can accelerate the development of efficacious treatment. Current studies on the RSV pathogenesis are based on the analysis of biopsies from the infected patients; however deeper understanding of molecular and cellular mechanisms of the RSV pathology could be achieved using animal models. Mice are the most often used model for RSV infection because they exhibit manifestations similar to those observed in humans (bronchial obstruction, mucous hypersecretion, and pulmonary inflammation mediated by lymphocytes, macrophages, and neutrophils). Additionally, the use of mice is economically feasible, and many molecular tools are available for studying RSV infection pathogenesis at the molecular and cellular levels. This review summarizes new data on the pathogenesis of RSV infection obtained in mouse models, which demonstrated the role of T cells in both the antiviral defense and the development of lung immunopathology. T cells not only eliminate the infected cells, but also produce significant amounts of the proinflammatory cytokines TNFα and IFNγ. Recently, a new subset of tissue-resident memory T cells (TRM) was identified that provide a strong antiviral defense without induction of lung immunopathology. These cells accumulate in the lungs after local rather than systemic administration of RSV antigens, which suggests new approaches to vaccination. The studies in mouse models have revealed a minor role of interferons in the anti-RSV protection, as RSV possesses mechanisms to escape the antiviral action of type I and III interferons, which may explain the low efficacy of interferon-containing drugs. Using knockout mice, a significant breakthrough has been achieved in understanding the role of many pro-inflammatory cytokines in lung immunopathology. It was found that in addition to TNFα and IFNγ, the cytokines IL-4, IL-5, IL-13, IL-17A, IL-33, and TSLP mediate the major manifestations of the RSV pathogenesis, such as bronchial obstruction, mucus hyperproduction, and lung infiltration by pro-inflammatory cells, while IL-6, IL-10, and IL-27 exhibit the anti-inflammatory effect. Despite significant differences between the mouse and human immune systems, mouse models have made a significant contribution to the understanding of molecular and cellular mechanisms of the pathology of human RSV infection.
Collapse
Affiliation(s)
- Igor P Shilovskiy
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia.
| | - Kirill V Yumashev
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Alexandr A Nikolsky
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Liudmila I Vishnyakova
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| | - Musa R Khaitov
- National Research Center, Institute of Immunology, Federal Medico-Biological Agency, Moscow, 115522, Russia
| |
Collapse
|
19
|
Anderson J, Do LAH, Wurzel D, Quan Toh Z, Mulholland K, Pellicci DG, Licciardi PV. Severe respiratory syncytial virus disease in preterm infants: a case of innate immaturity. Thorax 2021; 76:942-950. [PMID: 33574121 DOI: 10.1136/thoraxjnl-2020-216291] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/27/2021] [Indexed: 11/03/2022]
Abstract
Respiratory syncytial virus (RSV) is the most common viral pathogen associated with acute lower respiratory tract infection (LRTI) in children under 5 years of age. Severe RSV disease is associated with the development of chronic respiratory complications such as recurrent wheezing and asthma. A common risk factor for developing severe RSV disease is premature gestation and this is largely due to an immature innate immune system. This increases susceptibility to RSV since the innate immune system is less able to protect against pathogens at a time when adaptive immunity has not fully developed. This review focuses on comparing different aspects of innate immunity between preterm and term infants to better understand why preterm infants are more susceptible to severe RSV disease. Identifying early life innate immune biomarkers associated with the development of severe RSV disease, and understanding how these compare between preterm and term infants, remains a critically important question that would aid the development of interventions to reduce the burden of disease in this vulnerable population.
Collapse
Affiliation(s)
- Jeremy Anderson
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Lien Anh Ha Do
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| | - Danielle Wurzel
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia.,The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
| | - Zheng Quan Toh
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| | - Kim Mulholland
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia.,Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Daniel G Pellicci
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| | - Paul V Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia .,Department of Pediatrics, The University of Melbourne-Parkville Campus, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Eichinger KM, Kosanovich JL, Lipp M, Empey KM, Petrovsky N. Strategies for active and passive pediatric RSV immunization. Ther Adv Vaccines Immunother 2021; 9:2515135520981516. [PMID: 33623860 PMCID: PMC7879001 DOI: 10.1177/2515135520981516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 11/20/2020] [Indexed: 12/26/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children worldwide, with the most severe disease occurring in very young infants. Despite half a century of research there still are no licensed RSV vaccines. Difficulties in RSV vaccine development stem from a number of factors, including: (a) a very short time frame between birth and first RSV exposure; (b) interfering effects of maternal antibodies; and (c) differentially regulated immune responses in infants causing a marked T helper 2 (Th2) immune bias. This review seeks to provide an age-specific understanding of RSV immunity critical to the development of a successful pediatric RSV vaccine. Historical and future approaches to the prevention of infant RSV are reviewed, including passive protection using monoclonal antibodies or maternal immunization strategies versus active infant immunization using pre-fusion forms of RSV F protein antigens formulated with novel adjuvants such as Advax that avoid excess Th2 immune polarization.
Collapse
Affiliation(s)
- Katherine M. Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, and Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessica L. Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline Lipp
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kerry M. Empey
- Department of Pharmacy and Therapeutics, Department of Pharmaceutical Sciences, School of Medicine and Clinical and Translational Science Institute, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia 5042, Australia and Vaxine Pty Ltd, Warradale, SA 5046, Australia
| |
Collapse
|
21
|
Malinczak CA, Parolia A, Fonseca W, Morris S, Rasky AJ, Bawa P, Zhang Y, Mire MM, Ziegler SF, Ptaschinski C, Chinnaiyan AM, Lukacs NW. TSLP-Driven Chromatin Remodeling and Trained Systemic Immunity after Neonatal Respiratory Viral Infection. THE JOURNAL OF IMMUNOLOGY 2021; 206:1315-1328. [PMID: 33514510 DOI: 10.4049/jimmunol.2001205] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022]
Abstract
Our studies have previously shown a role for persistent TSLP production in the lungs of mice after early-life respiratory syncytial virus (RSV) infection that leads to an altered immune phenotype, including accumulation of "inflammatory" dendritic cells (DC). This study investigates the role of TSLP driving systemic trained immunity in DC in early-life RSV-infected mice. Bone marrow-derived DCs (BMDC) from early-life RSV-infected mice at 4 wk postinfection showed enhanced expression of costimulatory molecules and cytokines, including Tslp, that regulate immune cell function. The adoptive transfer of BMDC grown from early-life RSV-infected mice was sufficient to exacerbate allergic disease development. The addition of recombinant TSLP during differentiation of BMDC from naive mice induced a similar altered phenotype as BMDC grown from early-life RSV-infected mice, suggesting a role for TSLP in the phenotypic changes. To assess the role of TSLP in these changes, global transcriptomic characterization of TSLPR-/- BMDC infected with RSV was performed and showed a higher upregulation of type 1 IFN genes and concomitant downregulation of inflammatory genes. Assay for transposase-accessible chromatin using sequencing analysis demonstrated that TSLPR-/- BMDC had a parallel gain in physical chromatin accessibility near type 1 genes and loss in accessibility near genes related to RSV pathology, with IFN regulatory factor 4 (IRF4) and STAT3 predicted as top transcription factors binding within differentially accessible regions in wild-type. Importantly, these studies show that in the absence of TSLP signaling, BMDC are able to mount an appropriate type 1 IFN-associated antiviral response to RSV. In summary, RSV-induced TSLP alters chromatin structure in DC to drive trained innate immunity and activates pathogenic gene programs in mice.
Collapse
Affiliation(s)
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Susan Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Pushpinder Bawa
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Yuping Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Mohamed M Mire
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Steven F Ziegler
- Department of Immunology, Benaroya Research Institute, Seattle, WA 98101
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI 48109; and.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109.,Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
22
|
Makrinioti H, Custovic A, Hasegawa K, Camargo CA, Jartti T. The role of interferons in preschool wheeze. THE LANCET. RESPIRATORY MEDICINE 2021; 9:9-11. [PMID: 33412119 DOI: 10.1016/s2213-2600(20)30569-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 01/19/2023]
Affiliation(s)
- Heidi Makrinioti
- West Middlesex Hospital, Chelsea, and Westminster Foundation Trust, UK.
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, UK
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tuomas Jartti
- Department of Pediatrics, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
23
|
Control of IFN-I responses by the aminopeptidase IRAP in neonatal C57BL/6 alveolar macrophages during RSV infection. Mucosal Immunol 2021; 14:949-962. [PMID: 33846534 PMCID: PMC8221999 DOI: 10.1038/s41385-021-00402-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 03/03/2021] [Accepted: 03/22/2021] [Indexed: 02/04/2023]
Abstract
Respiratory Syncytial Virus (RSV) is the major cause of lower respiratory tract infection in infants, in whom, the sensing of RSV by innate immune receptors and its regulation are still poorly described. However, the severe bronchiolitis following RSV infection in neonates has been associated with a defect in type I interferons (IFN-I) production, a cytokine produced mainly by alveolar macrophages (AMs) upon RSV infection in adults. In the present study, neonatal C57BL/6 AMs mobilized very weakly the IFN-I pathway upon RSV infection in vitro and failed to restrain virus replication. However, IFN-I productions by neonatal AMs were substantially increased by the deletion of Insulin-Responsive AminoPeptidase (IRAP), a protein previously involved in the regulation of IFN-I production by dendritic cells. Moreover, neonatal IRAPKO AMs showed a higher expression of IFN-stimulated genes than their wild-type C57BL/6 counterpart. Interestingly, depletion of IRAP did not affect adult AM responses. Finally, we demonstrated that newborn IRAPKO mice infected with RSV had more IFN-I in their lungs and eliminated the virus more efficiently than WT neonates. Taken together, early-life susceptibility to RSV infection may be related to an original age-dependent suppressive function of IRAP on the IFN-I driven-antiviral responses in neonatal AMs.
Collapse
|
24
|
Jin L, Hu Q, Hu Y, Chen Z, Liao W. Respiratory Syncytial Virus Infection Reduces Kynurenic Acid Production and Reverses Th17/Treg Balance by Modulating Indoleamine 2,3-Dioxygenase (IDO) Molecules in Plasmacytoid Dendritic Cells. Med Sci Monit 2020; 26:e926763. [PMID: 33262321 PMCID: PMC7720431 DOI: 10.12659/msm.926763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) infection causes a world-wide medical and economic burden. This study analyzed the effects of RSV infection on plasmacytoid dendritic cells (pDCs) and evaluated the immunopathogenesis of RSV infection by measuring relative numbers of FoxP3+ Treg cells and Th17 cells. Material/Methods pDCs were isolated from human blood samples, purified using magnetic microbeads, and treated with RSV, IFN-γ, or vehicle. These cells were mixed with purified CD4+ T cells to yield preparations of pDCs+T cells+vehicle, pDCs+T cells+RSV, and pDCs+T cells+IFN-γ. Preparations of pDCs+T cells+RSV were also incubated with an inducer or an inhibitor of indoleamine 2,3-dioxygenase (IDO). Kynurenic acid concentration was measured by high-pressure liquid chromatography (HPLC). The differentiation of Foxp3+ Treg and Th17 cells from CD4+ T cells was determined by flow cytometry. Results pDCs were successfully isolated and purified using the magnetic microbeads. Compared with preparations of pDCs+T cells+vehicle, RSV infection (pDCs+T cells+RSV) significantly reduced and IFN-γ treatment (pDC+T cells+IFN-γ) increased kynurenic acid concentrations and the proportions of Foxp3+ Tregs (p<0.05 each). Conversely, RSV infection increased and IFN-γ treatment decreased the proportions of Th17 cells (p<0.05 each). RSV infection reduced kynurenic acid concentrations and inhibited the transformation from Th17 to Foxp3+ Tregs by modulating IDO molecules. Conclusions RSV infection reduced the production of kynurenic acid and inhibited transformation from Th17 to Foxp3+ Tregs (Th17/Treg balance) by modulating IDO molecules in pDCs.
Collapse
Affiliation(s)
- Ling Jin
- Department of Pediatrics, The First Affiliated Hospital (Southwest Hospital) of Army Medical University, Chongqing, China (mainland)
| | - Qi Hu
- Department of Pediatrics, The First Affiliated Hospital (Southwest Hospital) of Army Medical University, Chongqing, China (mainland)
| | - Ying Hu
- Department of Pediatrics, The First Affiliated Hospital (Southwest Hospital) of Army Medical University, Chongqing, China (mainland)
| | - Zhiqiang Chen
- Department of Pediatrics, The First Affiliated Hospital (Southwest Hospital) of Army Medical University, Chongqing, China (mainland)
| | - Wei Liao
- Department of Pediatrics, The First Affiliated Hospital (Southwest Hospital) of Army Medical University, Chongqing, China (mainland)
| |
Collapse
|
25
|
He L, Yang L, Zhang H, Luo Q. Efficacy and safety of interferon on neonates with respiratory syncytial virus pneumonia. Exp Ther Med 2020; 20:220. [PMID: 33193835 PMCID: PMC7646691 DOI: 10.3892/etm.2020.9350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) pneumonia is a leading cause of hospitalization and mortality among neonates worldwide, and there are currently no specific clinical treatments for RSV infection. Interferons (IFNs) possess broad-spectrum antiviral properties, and the present study aimed to evaluate the efficacy and safety of IFN-α1b for the treatment of neonatal RSV pneumonia. Neonates with RSV pneumonia were divided into the treatment (126 neonates) and control (160 neonates) groups, the former of which were treated with IFN. Aside from IFN administration, both groups received the same routine treatments. There were no significant differences in patient characteristics between the two groups. All neonates in the two groups displayed symptoms such as a cough (93.0%), tachypnea (90.1%), perilabial cyanosis (67.8%), choking on milk (62.9%) and moist rales (58.4%), and no significant differences in the occurrence of these symptoms were observed between the groups (P>0.05). The percentage of cases with bacterial co-infection was 66.8% (191/286), and the bacterial species in the spectrum primarily included Escherichia coli (21.5%), Klebsiella pneumonia (20.4%), Staphylococcus aureus (17.2%), Acihetobacter baumanii (13.1%) and Pseudomonas aeruginosa (9.9%). There were no significant differences in the co-infection rate or bacterial spectrum between the two groups. The remission time of cough, tachypnea, choking on milk, perilabial cyanosis, moist rales and oxygen inhalation in the treatment group was significantly lower compared with the control group (P<0.05). Although the hospitalization time in the treatment group was shorter compared with the control group, the difference was not significant. There were two patients in the treatment group that developed fever within 2-6 h after receiving IFN-α1b, though no other adverse effects were observed. In conclusion, IFN-α1b treatment improved the symptoms associated with neonatal RSV pneumonia with minimal adverse effects.
Collapse
Affiliation(s)
- Lingyun He
- Scientific Research and Education Section, Chongqing Health Center for Women and Children, Chongqing 401120, P.R. China
- Neonatal Department, Children's Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Lu Yang
- Department of Breast and Thyriod Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Hua Zhang
- Scientific Research and Education Section, Chongqing Health Center for Women and Children, Chongqing 401120, P.R. China
| | - Qian Luo
- Scientific Research and Education Section, Chongqing Health Center for Women and Children, Chongqing 401120, P.R. China
| |
Collapse
|
26
|
Bikhet M, Morsi M, Hara H, Rhodes LA, Carlo WF, Cleveland D, Cooper DK, Iwase H. The immune system in infants: Relevance to xenotransplantation. Pediatr Transplant 2020; 24:e13795. [PMID: 32845539 PMCID: PMC7606572 DOI: 10.1111/petr.13795] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/10/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022]
Abstract
Despite the improvement in surgical interventions in the treatment of congenital heart disease, many life-threatening lesions (eg, hypoplastic left heart syndrome) ultimately require transplantation. However, there is a great limitation in the availability of deceased human cardiac donors of a suitable size. Hearts from genetically engineered pigs may provide an alternative source. The relatively immature immune system in infants (eg, absence of anti-carbohydrate antibodies, reduced complement activation, reduced innate immune cell activity) should minimize the risk of early antibody-mediated rejection of a pig graft. Additionally, recipient thymectomy, performed almost routinely as a preliminary to orthotopic heart transplantation in this age-group, impairs the T-cell response. Because of the increasing availability of genetically engineered pigs (eg, triple-knockout pigs that do not express any of the three known carbohydrate antigens against which humans have natural antibodies) and the ability to diagnose congenital heart disease during fetal life, cardiac xenotransplantation could be preplanned to be carried out soon after birth. Because of these several advantages, prolonged graft survival and even the induction of tolerance, for example, following donor-specific pig thymus transplantation, are more likely to be achieved in infants than in adults. In this review, we summarize the factors in the infant immune system that would be advantageous in the success of cardiac xenotransplantation in this age-group.
Collapse
Affiliation(s)
- Mohamed Bikhet
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, AL, USA
| | - Mahmoud Morsi
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, AL, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, AL, USA
| | - Leslie A. Rhodes
- Division of Pediatric Cardiology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Waldemar F. Carlo
- Division of Pediatric Cardiology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David Cleveland
- Department of Pediatric Cardiovascular Surgery, Children’s Hospital of Alabama, Birmingham, AL, USA
| | - David K.C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, AL, USA
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
27
|
Challenges for the Newborn Immune Response to Respiratory Virus Infection and Vaccination. Vaccines (Basel) 2020; 8:vaccines8040558. [PMID: 32987691 PMCID: PMC7712002 DOI: 10.3390/vaccines8040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
The initial months of life reflect an extremely challenging time for newborns as a naïve immune system is bombarded with a large array of pathogens, commensals, and other foreign entities. In many instances, the immune response of young infants is dampened or altered, resulting in increased susceptibility and disease following infection. This is the result of both qualitative and quantitative changes in the response of multiple cell types across the immune system. Here we provide a review of the challenges associated with the newborn response to respiratory viral pathogens as well as the hurdles and advances for vaccine-mediated protection.
Collapse
|
28
|
Besteman SB, Callaghan A, Langedijk AC, Hennus MP, Meyaard L, Mokry M, Bont LJ, Calis JJA. Transcriptome of airway neutrophils reveals an interferon response in life-threatening respiratory syncytial virus infection. Clin Immunol 2020; 220:108593. [PMID: 32920212 DOI: 10.1016/j.clim.2020.108593] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Neutrophils are the most abundant cell type infiltrating the airways during severe respiratory syncytial virus (RSV) infection. Their exact role in disease pathophysiology remains enigmatic. Therefore, we determined genome-wide RNA expression profiles of local and systemic neutrophils in RSV bronchiolitis to provide further insight into local neutrophil biology. METHODS We performed a single-center analysis, in 16 infants, admitted to the pediatric intensive care unit with severe RSV bronchiolitis. Neutrophils were isolated from blood and tracheobronchial aspirates (sputum). After low input RNA sequencing, differential expression of genes was determined followed by gene set analysis. RESULTS Paired transcriptomic analysis of airway versus blood neutrophils showed an inflammatory phenotype, characterized by NF-kB signaling and upregulated expression of IL-6 and interferon pathways. We observed distinct expression of neutrophil activation genes (TNFSF13B, FCER1G). DISCUSSION Our data indicate that airway neutrophils regulate their function at the transcriptional level in response to viral infection. It also suggests that local interferon drives the neutrophil response of severe RSV bronchiolitis.
Collapse
Affiliation(s)
- Sjanna B Besteman
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Amie Callaghan
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Annefleur C Langedijk
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Marije P Hennus
- Department of Paediatric Intensive Care, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Oncode Institute, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Michal Mokry
- Department of cardiology, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Louis J Bont
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Jorg J A Calis
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands; Department of cardiology, University Medical Centre Utrecht, Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands.
| |
Collapse
|
29
|
Laubreton D, Drajac C, Eléouët JF, Rameix-Welti MA, Lo-Man R, Riffault S, Descamps D. Regulatory B Lymphocytes Colonize the Respiratory Tract of Neonatal Mice and Modulate Immune Responses of Alveolar Macrophages to RSV Infection in IL-10-Dependant Manner. Viruses 2020; 12:v12080822. [PMID: 32751234 PMCID: PMC7472339 DOI: 10.3390/v12080822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/11/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the prevalent pathogen of lower respiratory tract infections in children. The presence of neonatal regulatory B lymphocytes (nBreg) has been associated with a poor control of RSV infection in human newborns and with bronchiolitis severity. So far, little is known about how nBreg may contribute to neonatal immunopathology to RSV. We tracked nBreg in neonatal BALB/c mice and we investigated their impact on lung innate immunity, especially their crosstalk with alveolar macrophages (AMs) upon RSV infection. We showed that the colonization by nBreg during the first week of life is a hallmark of neonatal lung whereas this population is almost absent in adult lung. This particular period of age when nBreg are abundant corresponds to the same period when RSV replication in lungs fails to generate a type-I interferons (IFN-I) response and is not contained. When neonatal AMs are exposed to RSV in vitro, they produce IFN-I that in turn enhances IL-10 production by nBreg. IL-10 reciprocally can decrease IFN-I secretion by AMs. Thus, our work identified nBreg as an important component of neonatal lungs and pointed out new immunoregulatory interactions with AMs in the context of RSV infection.
Collapse
Affiliation(s)
- Daphné Laubreton
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (D.L.); (C.D.); (J.-F.E.)
| | - Carole Drajac
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (D.L.); (C.D.); (J.-F.E.)
| | - Jean-François Eléouët
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (D.L.); (C.D.); (J.-F.E.)
| | - Marie-Anne Rameix-Welti
- Université Paris-Saclay, UVSQ, Inserm, Infection et Inflammation, U1173, 78180 Montigny-Le-Bretonneux, France;
- Laboratoire de Microbiologie, Hôpital Ambroise Paré, AP-HP, 92100 Boulogne-Billancourt, France
| | - Richard Lo-Man
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Unit Immunity and Pediatric Infectious Diseases, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China;
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Sabine Riffault
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (D.L.); (C.D.); (J.-F.E.)
- Correspondence: (S.R.); (D.D.); Tel.: +(33)-01-34-65-26-20 (S.R.); +(33)-01-34-65-26-10 (D.D.)
| | - Delphyne Descamps
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (D.L.); (C.D.); (J.-F.E.)
- Correspondence: (S.R.); (D.D.); Tel.: +(33)-01-34-65-26-20 (S.R.); +(33)-01-34-65-26-10 (D.D.)
| |
Collapse
|
30
|
Kotomina T, Isakova-Sivak I, Stepanova E, Mezhenskaya D, Matyushenko V, Prokopenko P, Sivak K, Kiseleva I, Rudenko L. Neutralizing epitope of the Fusion Protein of Respiratory Syncytial Virus Embedded in the HA Molecule of LAIV Virus is not Sufficient to Prevent RS Virus Pulmonary Replication but Ameliorates Lung Pathology following RSV Infection in Mice. Open Microbiol J 2020. [DOI: 10.2174/1874285802014010147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aims:
To develop experimental bivalent vaccines against influenza and RSV using a cold-adapted LAIV backbone.
Background:
Respiratory syncytial virus (RSV) is a causative agent of bronchiolitis and pneumonia in young children, elderly and immunocompromised adults. No vaccine against RSV has been licensed to date for various reasons. One of the promising platforms for designing RSV vaccine is the use of live attenuated influenza vaccine (LAIV) viruses to deliver RSV epitopes to the respiratory mucosa.
Objective:
To generate recombinant LAIV viruses encoding a neutralizing epitope of the RSV fusion protein and assess their protective potential against both influenza and RSV infections in a mouse model.
Methods:
Reverse genetics methods were used to rescue recombinant LAIV+HA/RSV viruses expressing chimeric hemagglutinins encoding the RSV-F epitope at its N-terminus using two different flexible linkers. BALB/c mice were intranasally immunized with two doses of the recombinant viruses and then challenged with the influenza virus or RSV. The LAIV viral vector and formalin-inactivated RSV (FI-RSV) were included as control vaccines. Protection was assessed by the reduction of virus pulmonary titers. In addition, RSV-induced lung pathology was evaluated by histopathology studies.
Results:
Two rescued chimeric LAIV+HA/RSV viruses were identical to the LAIV vector in terms of replication capacity in vitro and in vivo. The RSV-F neutralizing epitope was successfully expressed only if inserted into the HA molecule via G-linker, but not A-linker. Both chimeric viruses induced high influenza-specific antibody levels and fully protected mice against a lethal influenza challenge virus. However, they induced weak anti-RSV antibody responses which did not prevent RS virus replication upon challenge, and only LAIV-HA+G-RSV variant protected mice against RSV-induced lung pathology.
Conclusion:
Although the designed LAIV-RSV chimeric viruses were unable to neutralize the RS virus pulmonary replication, the LAIV-HA+G-RSV reduced RSV-induced lung pathology and can be considered a promising bivalent vaccine against influenza and RSV infections and warrants its further development.
Collapse
|
31
|
Andrade CA, Pacheco GA, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM. Innate Immune Components that Regulate the Pathogenesis and Resolution of hRSV and hMPV Infections. Viruses 2020; 12:E637. [PMID: 32545470 PMCID: PMC7354512 DOI: 10.3390/v12060637] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.
Collapse
Affiliation(s)
- Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Gaspar A. Pacheco
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Nicolas M. S. Gálvez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| |
Collapse
|
32
|
Early-Life Respiratory Syncytial Virus Infection, Trained Immunity and Subsequent Pulmonary Diseases. Viruses 2020; 12:v12050505. [PMID: 32375305 PMCID: PMC7290378 DOI: 10.3390/v12050505] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) is often the first clinically relevant pathogen encountered in life, with nearly all children infected by two years of age. Many studies have also linked early-life severe respiratory viral infection with more pathogenic immune responses later in life that lead to pulmonary diseases like childhood asthma. This phenomenon is thought to occur through long-term immune system alterations following early-life respiratory viral infection and may include local responses such as unresolved inflammation and/or direct structural or developmental modifications within the lung. Furthermore, systemic responses that could impact the bone marrow progenitors may be a significant cause of long-term alterations, through inflammatory mediators and shifts in metabolic profiles. Among these alterations may be changes in transcriptional and epigenetic programs that drive persistent modifications throughout life, leaving the immune system poised toward pathogenic responses upon secondary insult. This review will focus on early-life severe RSV infection and long-term alterations. Understanding these mechanisms will not only lead to better treatment options to limit initial RSV infection severity but also protect against the development of childhood asthma linked to severe respiratory viral infections.
Collapse
|
33
|
Stephens LM, Varga SM. Function and Modulation of Type I Interferons during Respiratory Syncytial Virus Infection. Vaccines (Basel) 2020; 8:vaccines8020177. [PMID: 32290326 PMCID: PMC7349809 DOI: 10.3390/vaccines8020177] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory infections in infants and young children, accounting for an estimated 3 million hospitalizations annually worldwide. Despite the major health burden, there is currently no licensed RSV vaccine. RSV is recognized by a range of cellular receptors including both toll-like receptors (TLR) and retinoic acid-inducible gene-I-like receptors (RIG-I). This interaction initiates signaling through mitochondrial antiviral signaling (MAVS) and interferon regulatory factor (IRF) proteins, resulting in the induction of type I interferons (IFN). Early viral control is mediated by either IFN-α or IFN-β signaling through the IFN receptor (IFNAR), inducing the production of antiviral interferon-stimulating genes (ISGs). Type I IFNs also initiate the early production of proinflammatory cytokines including interleukin 6 (IL-6), tumor necrosis factor (TNF), and IFN-γ. Type I IFN levels correlate with age, and inadequate production may be a critical factor in facilitating the increased RSV disease severity observed in infants. Here, we review the current literature on the function of type I IFNs in RSV pathogenesis, as well as their involvement in the differential immune responses observed in infants and adults.
Collapse
Affiliation(s)
- Laura M. Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven M. Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
- Correspondence: ; Tel.: +1-319-335-7784
| |
Collapse
|
34
|
Type I IFN ineffectively activates neonatal dendritic cells limiting respiratory antiviral T-cell responses. Mucosal Immunol 2020; 13:371-380. [PMID: 31797910 PMCID: PMC7044048 DOI: 10.1038/s41385-019-0234-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/04/2023]
Abstract
Insufficient T-cell responses contribute to the increased burden of viral respiratory disease in infancy. Neonatal dendritic cells (DCs) often provide defective activation of pathogen-specific T cells through mechanisms that are incompletely understood, which hinders vaccine design for this vulnerable age group. Enhancing our characterization of neonatal DC sub-specialization and function is therefore critical to developing their potential for immunomodulation of T-cell responses. In this study, we engineered respiratory syncytial virus (RSV) to express a model protein, ovalbumin, to track antigen-presenting DCs in vivo. We found that murine neonatal conventional DC1s (cDC1s) efficiently migrated and presented RSV-derived antigen, challenging the paradigm that neonatal DCs are globally immature. In a key observation, however, we discovered that during infection neonatal cDC1s presenting viral antigen were unable to upregulate costimulatory molecules in response to type I interferons (IFN-I), contributing to poor antiviral T-cell responses. Importantly, we showed that the deficient response to IFN-I was also exhibited by human neonatal cDC1s, independent of infection. These findings reveal a functionally distinct response to IFN-I by neonatal cDC1s that may leave young infants susceptible to viral infections, and provide a new target for exploration, in light of failed efforts to design neonatal RSV vaccines.
Collapse
|
35
|
Contribution of Dendritic Cells in Protective Immunity against Respiratory Syncytial Virus Infection. Viruses 2020; 12:v12010102. [PMID: 31952261 PMCID: PMC7020095 DOI: 10.3390/v12010102] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe respiratory disease in infants and the elderly. The socioeconomic burden of RSV infection is substantial because it leads to serious respiratory problems, subsequent hospitalization, and mortality. Despite its clinical significance, a safe and effective vaccine is not yet available to prevent RSV infection. Upon RSV infection, lung dendritic cells (DCs) detecting pathogens migrate to the lymph nodes and activate the adaptive immune response. Therefore, RSV has evolved various immunomodulatory strategies to inhibit DC function. Due to the capacity of RSV to modulate defense mechanisms in hosts, RSV infection results in inappropriate activation of immune responses resulting in immunopathology and frequent reinfection throughout life. This review discusses how DCs recognize invading RSV and induce adaptive immune responses, as well as the regulatory mechanisms mediated by RSV to disrupt DC functions and ultimately avoid host defenses.
Collapse
|
36
|
Wu M, Gao L, He M, Liu H, Jiang H, Shi K, Shang R, Liu B, Gao S, Chen H, Gong F, Gelfand EW, Huang Y, Han J. Plasmacytoid dendritic cell deficiency in neonates enhances allergic airway inflammation via reduced production of IFN-α. Cell Mol Immunol 2019; 17:519-532. [PMID: 31853001 DOI: 10.1038/s41423-019-0333-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 11/09/2022] Open
Abstract
Allergic asthma, a chronic inflammatory airway disease associated with type 2 cytokines, often originates in early life. Immune responses at an early age exhibit a Th2 cell bias, but the precise mechanisms remain elusive. Plasmacytoid dendritic cells (pDCs), which play a regulatory role in allergic asthma, were shown to be deficient in neonatal mice. We report here that this pDC deficiency renders neonatal mice more susceptible to severe allergic airway inflammation than adult mice in an OVA-induced experimental asthma model. Adoptive transfer of pDCs or administration of IFN-α to neonatal mice prevented the development of allergic inflammation in wild type but not in IFNAR1-/- mice. Similarly, adult mice developed more severe allergic inflammation when pDCs were depleted. The protective effects of pDCs were mediated by the pDC-/IFN-α-mediated negative regulation of the secretion of epithelial cell-derived CCL20, GM-CSF, and IL-33, which in turn impaired the recruitment of cDC2 and ILC2 cells to the airway. In asthmatic patients, the percentage of pDCs and the level of IFN-α were lower in children than in adults. These results indicate that impairment of pDC-epithelial cell crosstalk in neonates is a susceptibility factor for the development of allergen-induced allergic airway inflammation.
Collapse
Affiliation(s)
- Min Wu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liuchuang Gao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Miao He
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hangyu Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Jiang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ketai Shi
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runshi Shang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Liu
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shan Gao
- Department of Respiratory Diseases, Wuhan Central Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hebin Chen
- Department of Pulmonary Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyan Han
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
37
|
Galeas-Pena M, McLaughlin N, Pociask D. The role of the innate immune system on pulmonary infections. Biol Chem 2019; 400:443-456. [PMID: 29604208 DOI: 10.1515/hsz-2018-0304] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/19/2018] [Indexed: 12/15/2022]
Abstract
Inhalation is required for respiration and life in all vertebrates. This process is not without risk, as it potentially exposes the host to environmental pathogens with every breath. This makes the upper respiratory tract one of the most common routes of infection and one of the leading causes of morbidity and mortality in the world. To combat this, the lung relies on the innate immune defenses. In contrast to the adaptive immune system, the innate immune system does not require sensitization, previous exposure or priming to attack foreign particles. In the lung, the innate immune response starts with the epithelial barrier and mucus production and is reinforced by phagocytic cells and T cells. These cells are vital for the production of cytokines, chemokines and anti-microbial peptides that are critical for clearance of infectious agents. In this review, we discuss all aspects of the innate immune response, with a special emphasis on ways to target aspects of the immune response to combat antibiotic resistant bacteria.
Collapse
Affiliation(s)
- Michelle Galeas-Pena
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, 333 S. Liberty St., New Orleans, LA 70112, USA
| | - Nathaniel McLaughlin
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, 333 S. Liberty St., New Orleans, LA 70112, USA
| | - Derek Pociask
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, 333 S. Liberty St., New Orleans, LA 70112, USA
| |
Collapse
|
38
|
Kumova OK, Fike AJ, Thayer JL, Nguyen LT, Mell JC, Pascasio J, Stairiker C, Leon LG, Katsikis PD, Carey AJ. Lung transcriptional unresponsiveness and loss of early influenza virus control in infected neonates is prevented by intranasal Lactobacillus rhamnosus GG. PLoS Pathog 2019; 15:e1008072. [PMID: 31603951 PMCID: PMC6808501 DOI: 10.1371/journal.ppat.1008072] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/23/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
Respiratory viral infections contribute substantially to global infant losses and disproportionately affect preterm neonates. Using our previously established neonatal murine model of influenza infection, we demonstrate that three-day old mice are exceptionally sensitive to influenza virus infection and exhibit high mortality and viral load. Intranasal pre- and post-treatment of neonatal mice with Lactobacillus rhamnosus GG (LGG), an immune modulator in respiratory viral infection of adult mice and human preterm neonates, considerably improves neonatal mice survival after influenza virus infection. We determine that both live and heat-killed intranasal LGG are equally efficacious in protection of neonates. Early in influenza infection, neonatal transcriptional responses in the lung are delayed compared to adults. These responses increase by 24 hours post-infection, demonstrating a delay in the kinetics of the neonatal anti-viral response. LGG pretreatment improves immune gene transcriptional responses during early infection and specifically upregulates type I IFN pathways. This is critical for protection, as neonatal mice intranasally pre-treated with IFNβ before influenza virus infection are also protected. Using transgenic mice, we demonstrate that the protective effect of LGG is mediated through a MyD88-dependent mechanism, specifically via TLR4. LGG can improve both early control of virus and transcriptional responsiveness and could serve as a simple and safe intervention to protect neonates.
Collapse
Affiliation(s)
- Ogan K. Kumova
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Adam J. Fike
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Jillian L. Thayer
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Linda T. Nguyen
- Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Joshua Chang Mell
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Judy Pascasio
- Pathology, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Christopher Stairiker
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Leticia G. Leon
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter D. Katsikis
- Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alison J. Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States of America
- Pediatrics, Drexel University College of Medicine, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
39
|
Balan S, Saxena M, Bhardwaj N. Dendritic cell subsets and locations. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:1-68. [PMID: 31810551 DOI: 10.1016/bs.ircmb.2019.07.004] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) are a unique class of immune cells that act as a bridge between innate and adaptive immunity. The discovery of DCs by Cohen and Steinman in 1973 laid the foundation for DC biology, and the advances in the field identified different versions of DCs with unique properties and functions. DCs originate from hematopoietic stem cells, and their differentiation is modulated by Flt3L. They are professional antigen-presenting cells that patrol the environmental interphase, sites of infection, or infiltrate pathological tissues looking for antigens that can be used to activate effector cells. DCs are critical for the initiation of the cellular and humoral immune response and protection from infectious diseases or tumors. DCs can take up antigens using specialized surface receptors such as endocytosis receptors, phagocytosis receptors, and C type lectin receptors. Moreover, DCs are equipped with an array of extracellular and intracellular pattern recognition receptors for sensing different danger signals. Upon sensing the danger signals, DCs get activated, upregulate costimulatory molecules, produce various cytokines and chemokines, take up antigen and process it and migrate to lymph nodes where they present antigens to both CD8 and CD4 T cells. DCs are classified into different subsets based on an integrated approach considering their surface phenotype, expression of unique and conserved molecules, ontogeny, and functions. They can be broadly classified as conventional DCs consisting of two subsets (DC1 and DC2), plasmacytoid DCs, inflammatory DCs, and Langerhans cells.
Collapse
Affiliation(s)
- Sreekumar Balan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Mansi Saxena
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| |
Collapse
|
40
|
Cheemarla NR, Uche IK, McBride K, Naidu S, Guerrero-Plata A. In utero tobacco smoke exposure alters lung inflammation, viral clearance, and CD8+T-cell responses in neonatal mice infected with respiratory syncytial virus. Am J Physiol Lung Cell Mol Physiol 2019; 317:L212-L221. [DOI: 10.1152/ajplung.00338.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Maternal smoking during pregnancy and exposure of infants to cigarette smoke are strongly associated with adverse health effects in childhood including higher susceptibility to respiratory viral infections. Human respiratory syncytial virus (HRSV) is the most important cause of lower respiratory tract infection among young infants. Exacerbation of respiratory disease, including HRSV bronchiolitis and higher susceptibility to HRSV infection, is well correlated with previous smoke exposure. The mechanisms of recurrence and susceptibility to viral pathogens after passive smoke exposure are multifactorial and include alteration of the structural and immunologic host defenses. In this work, we used a well-established mouse model of in utero smoke exposure to investigate the effect of in utero smoke exposure in HRSV-induced pathogenesis. Sample analysis indicated that in utero exposure led to increased lung inflammation characterized by an increased influx of neutrophils to the airways of the infected mice and a delayed viral clearance. On the other hand, decreased HRSV-specific CD8+T-cell response was observed. These findings indicate that cigarette smoke exposure during pregnancy alters HRSV-induced disease as well as several aspects of the neonatal immune responses.
Collapse
Affiliation(s)
- Nagarjuna R. Cheemarla
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Ifeanyi K. Uche
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Kaitlin McBride
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Shan Naidu
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana
- Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
41
|
Tognarelli EI, Bueno SM, González PA. Immune-Modulation by the Human Respiratory Syncytial Virus: Focus on Dendritic Cells. Front Immunol 2019; 10:810. [PMID: 31057543 PMCID: PMC6478035 DOI: 10.3389/fimmu.2019.00810] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/26/2019] [Indexed: 12/23/2022] Open
Abstract
The human respiratory syncytial virus (hRSV) is the leading cause of pneumonia in infants and produces a significant burden in the elderly. It can also infect and produce disease in otherwise healthy adults and recurrently infect those previously exposed to the virus. Importantly, recurrent infections are not necessarily a consequence of antigenic variability, as described for other respiratory viruses, but most likely due to the capacity of this virus to interfere with the host's immune response and the establishment of a protective and long-lasting immunity. Although some genes encoded by hRSV are known to have a direct participation in immune evasion, it seems that repeated infection is mainly given by its capacity to modulate immune components in such a way to promote non-optimal antiviral responses in the host. Importantly, hRSV is known to interfere with dendritic cell (DC) function, which are key cells involved in establishing and regulating protective virus-specific immunity. Notably, hRSV infects DCs, alters their maturation, migration to lymph nodes and their capacity to activate virus-specific T cells, which likely impacts the host antiviral response against this virus. Here, we review and discuss the most important and recent findings related to DC modulation by hRSV, which might be at the basis of recurrent infections in previously infected individuals and hRSV-induced disease. A focus on the interaction between DCs and hRSV will likely contribute to the development of effective prophylactic and antiviral strategies against this virus.
Collapse
Affiliation(s)
- Eduardo I Tognarelli
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
42
|
Hijano DR, Vu LD, Kauvar LM, Tripp RA, Polack FP, Cormier SA. Role of Type I Interferon (IFN) in the Respiratory Syncytial Virus (RSV) Immune Response and Disease Severity. Front Immunol 2019; 10:566. [PMID: 30972063 PMCID: PMC6443902 DOI: 10.3389/fimmu.2019.00566] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract disease in children <2 years of age. Increased morbidity and mortality have been reported in high-risk patients, such as premature infants, patients with cardiac disease, and severely immune compromised patients. Severe disease is associated with the virulence of the virus as well as host factors specifically including the innate immune response. The role of type I interferons (IFNs) in the response to RSV infection is important in regulating the rate of virus clearance and in directing the character of the immune response, which is normally associated with protection and less severe disease. Two RSV non-structural proteins, NS1 and NS2, as well as the envelope G glycoprotein are known to suppress type I IFN production and a robust type I IFN response to RSV does not occur in human infants or neonatal mouse models of RSV infection. Additionally, presence of type I IFNs are associated with mild symptoms in infants and administration of IFN-α prior to infection of neonatal mice with RSV reduces immunopathology. This evidence has driven RSV prophylaxis and therapeutic efforts to consider strategies for enhancing type I IFN production.
Collapse
Affiliation(s)
- Diego R Hijano
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Luan D Vu
- Department of Biological Sciences, Louisiana State University and School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | | - Ralph A Tripp
- Department of Infectious Disease, University of Georgia, Athens, GA, United States
| | | | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University and School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
43
|
Reizis B. Plasmacytoid Dendritic Cells: Development, Regulation, and Function. Immunity 2019; 50:37-50. [PMID: 30650380 PMCID: PMC6342491 DOI: 10.1016/j.immuni.2018.12.027] [Citation(s) in RCA: 382] [Impact Index Per Article: 76.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) are a unique sentinel cell type that can detect pathogen-derived nucleic acids and respond with rapid and massive production of type I interferon. This review summarizes our current understanding of pDC biology, including transcriptional regulation, heterogeneity, role in antiviral immune responses, and involvement in immune pathology, particularly in autoimmune diseases, immunodeficiency, and cancer. We also highlight the remaining gaps in our knowledge and important questions for the field, such as the molecular basis of unique interferon-producing capacity of pDCs. A better understanding of cell type-specific positive and negative control of pDC function should pave the way for translational applications focused on this immune cell type.
Collapse
Affiliation(s)
- Boris Reizis
- Department of Pathology and Department of Medicine, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
44
|
Han M, Rajput C, Ishikawa T, Jarman CR, Lee J, Hershenson MB. Small Animal Models of Respiratory Viral Infection Related to Asthma. Viruses 2018; 10:E682. [PMID: 30513770 PMCID: PMC6316391 DOI: 10.3390/v10120682] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/21/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Respiratory viral infections are strongly associated with asthma exacerbations. Rhinovirus is most frequently-detected pathogen; followed by respiratory syncytial virus; metapneumovirus; parainfluenza virus; enterovirus and coronavirus. In addition; viral infection; in combination with genetics; allergen exposure; microbiome and other pathogens; may play a role in asthma development. In particular; asthma development has been linked to wheezing-associated respiratory viral infections in early life. To understand underlying mechanisms of viral-induced airways disease; investigators have studied respiratory viral infections in small animals. This report reviews animal models of human respiratory viral infection employing mice; rats; guinea pigs; hamsters and ferrets. Investigators have modeled asthma exacerbations by infecting mice with allergic airways disease. Asthma development has been modeled by administration of virus to immature animals. Small animal models of respiratory viral infection will identify cell and molecular targets for the treatment of asthma.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Tomoko Ishikawa
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Caitlin R Jarman
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Julie Lee
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
45
|
Hijano DR, Siefker DT, Shrestha B, Jaligama S, Vu LD, Tillman H, Finkelstein D, Saravia J, You D, Cormier SA. Type I Interferon Potentiates IgA Immunity to Respiratory Syncytial Virus Infection During Infancy. Sci Rep 2018; 8:11034. [PMID: 30038294 PMCID: PMC6056463 DOI: 10.1038/s41598-018-29456-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/09/2018] [Indexed: 01/11/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection is the most frequent cause of hospitalization in infants and young children worldwide. Although mucosal RSV vaccines can reduce RSV disease burden, little is known about mucosal immune response capabilities in children. Neonatal or adult mice were infected with RSV; a subset of neonatal mice received interferon alpha (IFN-α) (intranasal) prior to RSV infection. B cells, B cell activating factor (BAFF) and IgA were measured by flow cytometry. RSV specific IgA was measured in nasal washes. Nasal associated lymphoid tissue (NALT) and lungs were stained for BAFF and IgA. Herein, we show in a mouse model of RSV infection that IFN-α plays a dual role as an antiviral and immune modulator and age-related differences in IgA production upon RSV infection can be overcome by IFN-α administration. IFN-α administration before RSV infection in neonatal mice increased RSV-specific IgA production in the nasal mucosa and induced expression of the B-cell activating factor BAFF in NALT. These findings are important, as mucosal antibodies at the infection site, and not serum antibodies, have been shown to protect human adults from experimental RSV infection.
Collapse
Affiliation(s)
- Diego R Hijano
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - David T Siefker
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bishwas Shrestha
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Sridhar Jaligama
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Luan D Vu
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Heather Tillman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jordy Saravia
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dahui You
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA.
| |
Collapse
|
46
|
Kimura D, Saravia J, Jaligama S, McNamara I, Vu LD, Sullivan RD, Mancarella S, You D, Cormier SA. New mouse model of pulmonary hypertension induced by respiratory syncytial virus bronchiolitis. Am J Physiol Heart Circ Physiol 2018; 315:H581-H589. [PMID: 29906223 DOI: 10.1152/ajpheart.00627.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pulmonary hypertension (PH) has been observed in up to 75% of infants with moderate to severe respiratory syncytial virus (RSV) bronchiolitis and is associated with significant morbidity and mortality in infants with congenital heart disease. The purpose of the present study was to establish a mouse model of PH secondary to RSV bronchiolitis that mimics the disease etiology as it occurs in infants. Neonatal mice were infected with RSV at 5 days of age and then reinfected 4 wk later. Serum-free medium was administered to age-matched mice as a control. Echocardiography and right ventricular systolic pressure (RVSP) measurements via right jugular vein catheterization were conducted 5 and 6 days after the second infection, respectively. Peripheral capillary oxygen saturation monitoring did not indicate hypoxia at 2-4 days post-RSV infection, before reinfection, and at 2-7 days after reinfection. RSV-infected mice had significantly higher RVSP than control mice. Pulsed-wave Doppler recording of the pulmonary blood flow by echocardiogram demonstrated a significantly shortened pulmonary artery acceleration time and decreased pulmonary artery acceleration time-to-ejection time ratio in RSV-infected mice. Morphometry showed that RSV-infected mice exhibited a significantly higher pulmonary artery medial wall thickness and had an increased number of muscularized pulmonary arteries compared with control mice. These findings, confirmed by RVSP measurements, demonstrate the development of PH in the lungs of mice infected with RSV as neonates. This animal model can be used to study the pathogenesis of PH secondary to RSV bronchiolitis and to assess the effect of treatment interventions. NEW & NOTEWORTHY This is the first mouse model of respiratory syncytial virus-induced pulmonary hypertension, to our knowledge. This model will allow us to decipher molecular mechanisms responsible for the pathogenesis of pulmonary hypertension secondary to respiratory syncytial virus bronchiolitis with the use of knockout and/or transgenic animals and to monitor therapeutic effects with echocardiography.
Collapse
Affiliation(s)
- Dai Kimura
- Department of Pediatrics, University of Tennessee Health Science Center, Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Jordy Saravia
- Department of Pediatrics, University of Tennessee Health Science Center, Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee.,Department of Immunology, St. Jude Children's Research Hospital , Memphis, Tennessee
| | - Sridhar Jaligama
- Department of Pediatrics, University of Tennessee Health Science Center, Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee.,Battelle Life Science Research, Columbus, Ohio
| | - Isabella McNamara
- Department of Pediatrics, University of Tennessee Health Science Center, Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee.,Department of Health Research Methods, Evidence, and Impact, McMaster University , Hamilton, Ontario , Canada
| | - Luan D Vu
- Department of Pediatrics, University of Tennessee Health Science Center, Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee.,Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Ryan D Sullivan
- Department of Comparative Medicine, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Salvatore Mancarella
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Dahui You
- Department of Pediatrics, University of Tennessee Health Science Center, Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee
| | - Stephania A Cormier
- Department of Pediatrics, University of Tennessee Health Science Center, Children's Foundation Research Institute, Le Bonheur Children's Hospital , Memphis, Tennessee.,Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| |
Collapse
|
47
|
Human respiratory syncytial virus: pathogenesis, immune responses, and current vaccine approaches. Eur J Clin Microbiol Infect Dis 2018; 37:1817-1827. [PMID: 29876771 DOI: 10.1007/s10096-018-3289-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/22/2018] [Indexed: 10/14/2022]
Abstract
Respiratory syncytial virus continues to pose a serious threat to the pediatric populations worldwide. With a genomic makeup of 15,200 nucleotides, the virus encodes for 11 proteins serving as envelope spikes, inner envelope proteins, and non-structural and ribonucleocapsid complexes. The fusion (F) and attachment (G) surface glycoproteins are the key targets for neutralizing antibodies. The highly variable G with altered glycosylations and the conformational alternations of F create challenges for vaccine development. The metastable F protein is responsible for RSV-host cell fusion and thus infectivity. Novel antigenic sites were identified on this form following its stabilization and solving its crystal structure. Importantly, site ø displays neutralizing activity exceeding those of post-F-specific and shared antigenic sites, such as site II which is the target for Palivizumab therapeutic antibody. Induction of high neutralizing antibody responses by pre-F immunization in animal models promoted it as a major vaccine candidate. Since RSV infection is more serious at age extremities and in individuals with undermining health conditions, vaccines are being developed to target these populations. Infants below three months of age have a suppressive immune system, making vaccines' immunogenicity weak. Therefore, a suggested strategy to protect newborns from RSV infection would be through passive immunity of maternal antibodies. Hence, pregnant women at their third trimester have been selected as an ideal target for vaccination with RSV pre-F vaccine. This review summarizes the different modes of RSV pathogenesis and host's immune response to the infection, and illustrates on the latest updates of vaccine development and vaccination approaches.
Collapse
|
48
|
Restori KH, Srinivasa BT, Ward BJ, Fixman ED. Neonatal Immunity, Respiratory Virus Infections, and the Development of Asthma. Front Immunol 2018; 9:1249. [PMID: 29915592 PMCID: PMC5994399 DOI: 10.3389/fimmu.2018.01249] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/18/2018] [Indexed: 12/27/2022] Open
Abstract
Infants are exposed to a wide range of potential pathogens in the first months of life. Although maternal antibodies acquired transplacentally protect full-term neonates from many systemic pathogens, infections at mucosal surfaces still occur with great frequency, causing significant morbidity and mortality. At least part of this elevated risk is attributable to the neonatal immune system that tends to favor T regulatory and Th2 type responses when microbes are first encountered. Early-life infection with respiratory viruses is of particular interest because such exposures can disrupt normal lung development and increase the risk of chronic respiratory conditions, such as asthma. The immunologic mechanisms that underlie neonatal host-virus interactions that contribute to the subsequent development of asthma have not yet been fully defined. The goals of this review are (1) to outline the differences between the neonatal and adult immune systems and (2) to present murine and human data that support the hypothesis that early-life interactions between the immune system and respiratory viruses can create a lung environment conducive to the development of asthma.
Collapse
Affiliation(s)
- Katherine H Restori
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Bharat T Srinivasa
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Elizabeth D Fixman
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
49
|
Mebratu YA, Tesfaigzi Y. IL-17 Plays a Role in Respiratory Syncytial Virus-induced Lung Inflammation and Emphysema in Elastase and LPS-injured Mice. Am J Respir Cell Mol Biol 2018; 58:717-726. [PMID: 29314865 PMCID: PMC6002655 DOI: 10.1165/rcmb.2017-0265oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/09/2018] [Indexed: 01/01/2023] Open
Abstract
Respiratory syncytial virus (RSV) is associated with enhanced progression of chronic obstructive pulmonary disease (COPD) and COPD exacerbations. However, little is known about the role of IL-17 in RSV-induced lung injury. We first investigated the role of RSV infection in enhancing mucous cell hyperplasia (MCH) and airspace enlargement in the lungs of mice injured with elastase and LPS (E/LPS). Mice injured with E/LPS had an enhanced and prolonged neutrophilic response to RSV that was associated with decreased levels of type I IFN and increased levels of IL-17, IL-23, CXCL-1, granulocyte colony stimulating factor (GCSF), CXCL-5, and matrix metalloproteinase (MMP)-9. In addition, extent of MCH and mean weighted alveolar space were increased significantly in the lungs of E/LPS-injured mice infected with RSV compared with E/LPS-only or RSV-only controls. Interestingly, immunodepletion of IL-17 before viral infection diminished the RSV-driven MCH and airspace enlargement in the E/LPS-injured animals, suggesting that IL-17 may be a therapeutic target for MCH and airspace enlargement when enhanced by RSV infection.
Collapse
Affiliation(s)
- Yohannes A Mebratu
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Yohannes Tesfaigzi
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| |
Collapse
|
50
|
Ascough S, Paterson S, Chiu C. Induction and Subversion of Human Protective Immunity: Contrasting Influenza and Respiratory Syncytial Virus. Front Immunol 2018; 9:323. [PMID: 29552008 PMCID: PMC5840263 DOI: 10.3389/fimmu.2018.00323] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/06/2018] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) and influenza are among the most important causes of severe respiratory disease worldwide. Despite the clinical need, barriers to developing reliably effective vaccines against these viruses have remained firmly in place for decades. Overcoming these hurdles requires better understanding of human immunity and the strategies by which these pathogens evade it. Although superficially similar, the virology and host response to RSV and influenza are strikingly distinct. Influenza induces robust strain-specific immunity following natural infection, although protection by current vaccines is short-lived. In contrast, even strain-specific protection is incomplete after RSV and there are currently no licensed RSV vaccines. Although animal models have been critical for developing a fundamental understanding of antiviral immunity, extrapolating to human disease has been problematic. It is only with recent translational advances (such as controlled human infection models and high-dimensional technologies) that the mechanisms responsible for differences in protection against RSV compared to influenza have begun to be elucidated in the human context. Influenza infection elicits high-affinity IgA in the respiratory tract and virus-specific IgG, which correlates with protection. Long-lived influenza-specific T cells have also been shown to ameliorate disease. This robust immunity promotes rapid emergence of antigenic variants leading to immune escape. RSV differs markedly, as reinfection with similar strains occurs despite natural infection inducing high levels of antibody against conserved antigens. The immunomodulatory mechanisms of RSV are thus highly effective in inhibiting long-term protection, with disturbance of type I interferon signaling, antigen presentation and chemokine-induced inflammation possibly all contributing. These lead to widespread effects on adaptive immunity with impaired B cell memory and reduced T cell generation and functionality. Here, we discuss the differences in clinical outcome and immune response following influenza and RSV. Specifically, we focus on differences in their recognition by innate immunity; the strategies used by each virus to evade these early immune responses; and effects across the innate-adaptive interface that may prevent long-lived memory generation. Thus, by comparing these globally important pathogens, we highlight mechanisms by which optimal antiviral immunity may be better induced and discuss the potential for these insights to inform novel vaccines.
Collapse
Affiliation(s)
- Stephanie Ascough
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Suzanna Paterson
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| | - Christopher Chiu
- Section of Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| |
Collapse
|