Elsevier

Virus Research

Volume 167, Issue 2, August 2012, Pages 138-145
Virus Research

Review
Pattern recognition receptors for respiratory syncytial virus infection and design of vaccines

https://doi.org/10.1016/j.virusres.2012.06.003Get rights and content

Abstract

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract illness in infants and young children. Host immune response has been implicated in both the protection and immunopathological mechanisms. Pattern recognition receptors (PRRs) expressed on innate immune cells during RSV infection recognize the RSV-associated molecular patterns and activate innate immune cells as well as mediate airway inflammation, protective immune response, and pulmonary immunopathology. The resident and recruited innate immune cells play important roles in the protection and pathogenesis of an RSV disease by expressing these PRRs. Agonist-binding PRRs are the basis of many adjuvants that are essential for most vaccines. In the present review, we highlight recent advances in the innate immune recognition of and responses to RSV through PRRs, including toll-like receptors (TLRs), retinoic acid-inducible gene (RIG)-I-like receptors (RLRs), and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs). We also describe the role of PRRs in the design of RSV vaccines.

Introduction

Respiratory syncytial virus (RSV) is the most important cause of severe lower respiratory tract infection in infants and young children worldwide (Collins et al., 2001). Recent evidence suggests that RSV is an increasing cause of morbidity and mortality in the elderly and transplant patients as well as patients with chronic obstructive pulmonary disease (Thompson et al., 2003). Despite the importance of RSV as a respiratory pathogen, no licensed vaccines and effective therapy strategy are available, although palivizumab, a humanized monoclonal antibody, has been used for immunoprophylaxis against RSV in high-risk infants and young children (The IMpact-RSV Study Group, 1998). Understanding of the mechanisms that maintain the respiratory illness is limited (Collins and Melero, 2011). In-depth understanding of RSV-infection mechanisms and the host immune responses involved is essential to the development of effective vaccines. The host immune response to RSV has been implicated in both the protection and immunopathological mechanisms.

Successfully protecting the host from pathogens involves rapid activation of innate immune responses that serve as the first line of defense against invading pathogens and tailors the adaptive immune responses. Activation of innate immunity depends on the recognition of pathogen-associated molecular patterns (PAMPs) that are specific for the pathogen, but absent in the host using pattern recognition receptors (PRRs) expressed on sentinel cells. To date, three classes of PRRs have been identified, including toll-like receptors (TLRs), retinoic acid-inducible gene (RIG)-I-like receptors (RLRs), and nucleotide-binding oligomerization domain (NOD)-like receptor (NLRs) (Ohto et al., 2007, Kumar et al., 2009). TLRs on the cell surface and endosomal compartments recognize a wide range of PAMPs and play a central role in initiating innate immune responses. There are currently 11 known human TLRs (Leulier and Lemaitre, 2008). RLRs, such as RIG-I and MDA5, belong to the RNA helicase family and have been identified as essential cytosolic receptors for intracellular viral RNAs, mediating the anti-viral programs via type I IFN induction (Yoneyama et al., 2005). The NLRs constitute a large cytosolic receptor family. The NLR family members NOD1, NOD2, and NALP3 recognize PAMPs in the cytosol as well as play a role in innate immunity (Akira et al., 2006). The cytosolic NLR and RLR families contribute to the immune response against pathogens in collaboration with the membrane-bound TLRs (Yoneyama and Fujita T, 2008). All these PRRs recognize various PAMPs and activate transcription factors NF-κB, mitogen-activated protein kinases (MAPKs), and/or members of the interferon regulatory factor (IRF) family, which regulate the expression of inflammatory cytokines and type I interferons (Medzhitov, 2007). RSV can be recognized by these three classes of PRRs. Airway resident leukocytes, such as dendritic cells (DCs) and macrophages as well as recruited proinflammatory granulocytes (neutrophils and eosinophils), are markedly involved in mediating airway inflammation and asthma pathophysiology by expressing these PRRs during RSV infection (Amanatidou et al., 2009, Murawski et al., 2009). Adjuvants are essential for enhancing vaccine efficacy. Agonist-binding PRRs are the basis of many adjuvants. These molecules exert their adjuvant function by interacting with TLRs, NLRs, RLRs, and signal through MyD88-dependent and MyD88-independent pathways (Higgins and Mills, 2010). In the present review, we highlight recent advances in the innate immune recognition of and responses to RSV through PRRs, which resulted in appropriate antiviral responses and/or pulmonary immunopathology. We also describe the role of PRRs in the design of RSV vaccines.

Section snippets

TLR-mediated innate immune recognition of RSV

TLRs are found on a wide range of cells such as macrophages, DCs, epithelial cells, eosinophils, and neutrophils. TLR1, TLR2, TLR4, TLR5, and TLR6 reside on the cell surface as well as recognize microbial surface molecules and/or products, such as LPS or RSV fusion (F) protein. TLR3, TLR7, TLR8, and TLR9 are expressed in intracellular vesicles as well as recognize viral nucleic acids. Ligand recognition by TLRs, except TLR3, leads to the activation of the MyD88-dependent pathway, whereas TLR3

RLRs as sensors of RSV infection

RLRs are the critical sensors of viral infection in most cell types including macrophage and conventional DCs. RIG-I and MDA5 contain a DExD/H box RNA-binding helicase domain as well as two caspase activation and recruitment domains (CARDs) (Fujita et al., 2007). RIG-I is activated by 5’ triphosphate structure of viral RNA independently of single or double strand (Hornung et al., 2006). Scagnolari et al. (2009) reported that the relative gene expression levels of MDA5 and RIG-I were

NLRs involved in RSV recognition

NLRs consist of three domains characterized by an N-terminal effector and a central nucleotide-binding (NBD) domains as well as a C-terminal multiple leucine-rich repeats (LRR) (Ting et al., 2008). The NLR family is divided into five subfamilies according to their effector domain (Chen et al., 2009). NOD1 and NOD2 are well-characterized, which recognize distinct motifs in peptidoglycan structures of intracellular bacteria (Millmann-Born et al., 2010) as well as induce pro-inflammatory and

Innate immune cells respond to RSV infection by PRRs

Two major types of DCs, namely, myeloid/conventional (mDCs/cDCs) and plasmacytoid (pDCs), have been discovered. DCs are the major antigen-presenting cells following RSV infection (Lukens et al., 2009). RSV infects cDCs and mDCs, increasing surface expression of maturation markers as well as cytokine production (Johnson et al., 2011). RIG-I is expressed in both pDCs and cDC (Kato et al., 2005). NLR signaling enhances DC-mediated cross-priming in vivo (Asano et al., 2010). cDCs have a relatively

Design of RSV vaccines

Since triggering via PRR expressed on different innate immune cells induces host's immune response, PRR ligands can be harnessed to enhance the induction and regulate the types of vaccine-specific responses. Adjuvants are essential factors for the development of modern vaccines that promote various types of immune responses. The ability of agonists interacting with PRRs to modulate immunity provides a promising new tool for novel adjuvant development and contributes to understanding the

Conclusion

The binding of RSV surface proteins to TLR1, TLR2, TLR4, and TLR6, as well as RSV RNAs to TLR3, TLR7, RLRs, and NLRs activates important innate immune pathways. This phenomenon also promotes the expression of inflammatory cytokines and chemokines (which activate and/or recruit innate, as well as adaptive, immune cells), resulting in the clearance of RSV or immunopathology. Understanding the interaction between RSV and its host is crucial for the development of novel therapeutic strategies as

Acknowledgments

The present work was supported by grants from the Chinese National Natural Science Foundation (No. 30972801), the 100 Innovation Talents Project of Hebei Higher Education (CPRC030), and Specialized Research Fund for the Doctoral Program of Higher Education, State Education Ministry (No. 20091323120005).

References (108)

  • Y. Huang et al.

    Murine host responses to respiratory syncytial virus (RSV) following intranasal administration of a Protollin-adjuvanted, epitope-enhanced recombinant G protein vaccine

    Journal of Clinical Virology

    (2009)
  • T.R. Johnson et al.

    TLR9 agonist, but not TLR7/8, functions as an adjuvant to diminish FI-RSV vaccine-enhanced disease, while either agonist used as therapy during primary RSV infection increases disease severity

    Vaccine

    (2009)
  • H. Kato et al.

    Cell type-specific involvement of RIG-I in antiviral response

    Immunity

    (2005)
  • H. Kumar et al.

    Toll-like receptors and innate immunity

    Biochemical and Biophysical Research Communications

    (2009)
  • C. Le Nouën et al.

    Infection and maturation of monocyte-derived human dendritic cells by human respiratory syncytial virus, human metapneumovirus, and human parainfluenza virus type 3

    Virology

    (2009)
  • P. Liu et al.

    Respiratory syncytial virus induces RelA release from cytoplasmic 100-kDa NF-κB2 complexes via a novel retinoic acid-inducible gene-I·NF-κB-inducing kinase signaling pathway

    Journal of Biological Chemistry

    (2008)
  • K.H.G. Mills

    Designer adjuvants for enhancing the efficacy of infectious disease and cancer vaccines based on suppression of regulatory T cell induction

    Immunology Letters

    (2009)
  • M.M. Monick et al.

    Respiratory syncytial virus upregulates TLR4 and sensitizes airway epithelial cells to endotoxin

    Journal of Biological Chemistry

    (2003)
  • L.O. Moreira et al.

    Modulation of adaptive immunity by different adjuvant–antigen combinations in mice lacking Nod2

    Vaccine

    (2008)
  • S. Phipps et al.

    Eosinophils contribute to innate antiviral immunity and promote clearance of respiratory syncytial virus

    Blood

    (2007)
  • A.C. Schmidt et al.

    Respiratory syncytial virus and other pneumoviruses: a review of the international symposium-RSV 2003

    Virus Research

    (2004)
  • K.A. Shirey et al.

    Control of RSV-induced Lung Injury by Alternatively Activated Macrophages Is IL-4Rα-, TLR4-, and IFN-β-dependent

    Mucosal Immunology

    (2010)
  • S.R. Singh et al.

    Immunogenicity and efficacy of recombinant RSV-F vaccine in a mouse model

    Vaccine

    (2007)
  • T. Suzuki et al.

    Role of innate immune cells and their products in lung immunopathology

    International Journal of Biochemistry & Cell Biology

    (2008)
  • J.P. Ting et al.

    The NLR gene family: a standard nomenclature

    Immunity

    (2008)
  • F. Aeffner et al.

    Double-stranded RNA induces similar pulmonary dysfunction to respiratory syncytial virus in BALB/c mice

    American Journal of Physiology Lung Cellular and Molecular Physiology

    (2011)
  • V. Amanatidou et al.

    Genetic diversity of the host and severe respiratory syncytial virus-induced lower respiratory tract infection

    Pediatric Infectious Disease Journal

    (2009)
  • J. Asano et al.

    Nucleotide oligomerization binding domain-like receptor signaling enhances dendritic cell-mediated cross-priming in vivo

    Journal of Immunology

    (2010)
  • A.A. Awomoyi et al.

    Association of TLR4 polymorphisms with symptomatic respiratory syncytial virus infection in high-risk infants and young children

    Journal of Immunology

    (2007)
  • K. Banasal et al.

    Intracellular pathogen sensor NOD2 programs macrophages to trigger Notch1 activation

    The Journal of Biological Chemistry

    (2010)
  • V.G. Bhoj et al.

    MAVS and MyD88 are essential for innate immunity but not cytotoxic T lymphocyte response against respiratory syncytial virus

    Proceedings of the National Academy of Sciences

    (2008)
  • J.C.G. Blanco et al.

    New insights for development of a safe and protective RSV

    Vaccine: Human Vaccines

    (2010)
  • I. Boogaard et al.

    Respiratory syncytial virus differentially activates murine myeloid and plasmacytoid dendritic cells

    Immunology

    (2007)
  • K.N. Carroll et al.

    The severity-dependent relationship of infant bronchiolitis on the risk and morbidity of early childhood asthma

    Journal of Allergy and Clinical Immunology

    (2009)
  • A.M. Cepika et al.

    Expression of chemokine receptor CX3CR1 in infants with respiratory syncytial virus bronchiolitis

    Pediatric Allergy and Immunology

    (2008)
  • G. Chen et al.

    Nod-like receptors: role in innate immunity and inflammatory disease

    Annual Review of Pathology

    (2009)
  • J. Choi et al.

    The role of TNF-α in eosinophilic inflammation associated with RSV bronchiolitis

    Pediatric Allergy and Immunology

    (2010)
  • P.L. Collins et al.
  • M.F. Delgado et al.

    Lack of antibody affinity maturation due to poor Toll-like receptor stimulation leads to enhanced respiratory syncytial virus disease

    Nature Medicine

    (2009)
  • S.S. Diebold et al.

    Innate anti-viral responses via TLR7-mediated recognition of single stranded RNA

    Science

    (2004)
  • S. Ehl et al.

    The role of toll-like receptor 4 versus interleukin-12 in immunity to respiratory syncytial virus

    European Journal of Immunology

    (2004)
  • S.C. Eisenbarth et al.

    Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants

    Nature

    (2008)
  • A. Gagro et al.

    Increased Toll-like receptor 4 expression in infants with respiratory syncytial virus bronchiolitis

    Clinical and Experimental Immunology

    (2004)
  • D.J. Groskreutz et al.

    Respiratory syncytial virus induces TLR3 protein and protein kinase R, leading to increased double-stranded RNA responsiveness in airway epithelial cells

    Journal of Immunology

    (2006)
  • H.A. Haeberle et al.

    Respiratory syncytial virus-induced activation of nuclear factor-kappaB in the lung involves alveolar macrophages and toll-like receptor 4-dependent pathways

    Journal of Infectious Diseases

    (2002)
  • H.A. Haerberle et al.

    Respiratory syncytial virus-induced activation of nuclear factor-kappa B in the lung involves alveolar macrophages and Toll like receptor 4-dependant pathways

    Journal of Infectious Diseases

    (2002)
  • C.P. Halfhide et al.

    Neutrophil TLR4 expression is reduced in the airways of infants with severe bronchiolitis

    Thorax

    (2009)
  • L.M. Haynes et al.

    Involvement of toll-like receptor 4 in innate immunity to respiratory syncytial virus

    Journal of Virology

    (2001)
  • S.C. Higgins et al.

    TLR, NLR agonists, and other immune modulators as infectious disease vaccine adjuvants

    Current Infectious Diseases Reports

    (2010)
  • V. Hornung et al.

    50-Triphosphate RNA is the ligand for RIG-I

    Science

    (2006)
  • Cited by (32)

    • Topical Microbial Therapeutics against Respiratory Viral Infections

      2021, Trends in Molecular Medicine
      Citation Excerpt :

      Several PRRs can simultaneously mediate responses to viral and bacterial infections. For example, immune responses to the different RSV infection components are mediated by NOD2, RIG-I, and TLRs in the airways, including the extracellular TLR2/6 and TLR4 that recognize viral envelope proteins (e.g., the RSV fusion protein) [46] and endosomal TLR3 and TLR7 that recognize dsRNA and ssRNA, respectively [63]. The innate signaling cascades culminate in the active expression of proinflammatory and regulatory cytokines, IFNs, growth factors, proteases, antimicrobial peptides, reactive oxygen species, and mucus-related genes [24] (Figure 1).

    • Inflammatory damage on respiratory and nervous systems due to hRSV infection

      2015, Current Opinion in Immunology
      Citation Excerpt :

      hRSV recognition by AECs leads to the secretion of a broad spectrum of cytokines and chemokines [22]. These molecules promote the recruitment of cells to the peribronchiolar space, including eosinophils, neutrophils and monocytes, as well as the onset of an anti-viral response (Figure 1) [22,24]. A study, comparing nasal-derived and bronchial-derived epithelia from the same individuals, showed a similar pro-inflammatory chemokine response (CXCL10, CCL5, and CXCL11) in vitro upon hRSV infection [5•].

    • Contribution of autophagy to antiviral immunity

      2015, FEBS Letters
      Citation Excerpt :

      Toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) and Retinoic acid-inducible gene (RIG-1)-I-like receptors (RLRs) are the pattern recognition receptors (PRR) involved in detecting pathogen components, such as single- and double-stranded viral RNAs [45,46]. Several TLRs are known to activate innate responses after virus infection, including TLR2, TLR3, TLR4, TLR7 and TLR8 [45,46]. TLR2 and TLR4 are expressed on the cell surface whereas TLR3, TLR7 and TLR8 are localized within cytoplasmatic compartments, such as endosomes [47–49].

    • Co-immunization with virus-like particle and DNA vaccines induces protection against respiratory syncytial virus infection and bronchiolitis

      2014, Antiviral Research
      Citation Excerpt :

      It was also reported that there was a correlation between RSV protection and levels of F specific antibodies (Kasel et al., 1987). RSV F is known to stimulate Toll-like receptor 4 (Zeng et al., 2012), which might be contributing to higher immune responses to F compared to G in the groups of live RSV and FFG VLP. Taken together, these results in this study suggest that uniquely combined FFG VLP can induce long-lived IgG2a antibody responses specific for RSV F. Mice immunized with FFG VLP showed long term protection against RSV without causing lung disease.

    • Viral infections in asthma and COPD

      2014, Respiratory Investigation
      Citation Excerpt :

      RIG-I recognizes the 5'-triphosphate motif of RSV ssRNA as well as its dsRNA product [20], while MDA5 recognizes HRV dsRNA and poly IC. Additionally, HRV is recognized by RIG-I and TLR3 [20]. Both RIG-I and MDA5 initiate signaling through IFN-β promoter stimulator 1 (IPS-1), which is also known as MAVS/Cardif/VISA [21–24].

    View all citing articles on Scopus
    View full text