Immune Netw. 2013 Oct;13(5):163-167. English.
Published online Oct 26, 2013.
Copyright © 2013 The Korean Association of Immunologists
Review

Respiratory Syncytial Virus (RSV) Modulation at the Virus-Host Interface Affects Immune Outcome and Disease Pathogenesis

Ralph A. Tripp
    • Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens GA 30602, USA.
Received August 14, 2013; Revised August 23, 2013; Accepted August 28, 2013.

This is an open access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

The dynamics of the virus-host interface in the response to respiratory virus infection is not well-understood; however, it is at this juncture that host immunity to infection evolves. Respiratory viruses have been shown to modulate the host response to gain a replication advantage through a variety of mechanisms. Viruses are parasites and must co-opt host genes for replication, and must interface with host cellular machinery to achieve an optimal balance between viral and cellular gene expression. Host cells have numerous strategies to resist infection, replication and virus spread, and only recently are we beginning to understand the network and pathways affected. The following is a short review article covering some of the studies associated with the Tripp laboratory that have addressed how respiratory syncytial virus (RSV) operates at the virus-host interface to affects immune outcome and disease pathogenesis.

Keywords
RSV; Virus-host interface; Disease intervention

INTRODUCTION

People of all ages experience viral respiratory tract infections each year, with young children, the elderly, and immune compromised most severely afflicted (1). Epidemiological surveys of diagnostic studies have identified common agents that include respiratory syncytial virus (RSV), influenza viruses (flu), human metapneumovirus (HMPV), rhinoviruses (RV) parainfluenza virus (PIV), adenovirus (Ad), and human coronavirus (hCoV) (2-4). Common to these viruses are their ability to infect airway epithelial cells, co-opt host cell proteins to facilitate infection, modulate both innate and adaptive immune responses, and to mediate proinflammatory responses which contribute to disease pathogenesis. Despite the airways constantly being challenged, airway defense mechanisms generally protect from disease with minimal clinical consequences.

VIRUS-HOST INTERACTION

It is well-understood that airway epithelial cells possess innate immune functions that control infection and replication, and recruit, activate and facilitate expansion of adaptive immune responses to facilitate clearance of infected epithelial cells (5-7). This response is in part mediated by pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) that act as viral sensors of infection (8, 9). The distribution of TLRs has been shown to depend on the type of cells and their localization (10, 11). TLR4 has been shown to be important in sensing RSV infection and its expression is linked to disease outcome (12-14), however all viruses are sensed via PRRs. RNA from viruses such as influenza and RSV is rapidly detected during replication by intracellular PRRS such as RIG-I and MDA5 (15). These viruses induce activation of the IFN-promoter via RIG-I signaling, and this signaling has a role in the antiviral response to infection (16, 17).

RSV interaction with host airway epithelium leads to the induction cytokines, chemokines and antiviral agents. This response occurs very early, i.e. before the expression of viral proteins (7). For example, TLR4 is expressed on the cell surface of respiratory epithelial cells, and responds to the RSV fusion protein (12, 13). TLR4 signals through MyD88 to activate NF-kappa B, and via TRIF, to activate IRF-7, and cytokine, chemokine and IFN expression (18). The resulting cytokine cascade leads to a pro-inflammatory response that must be negatively regulated, primarily through suppressor of cytokine signaling (SOCS) proteins (19-21). Recently, a novel class of antiviral cytokines was discovered and classified as type III IFNs: IFN-lambda1 (IL-29), IFN-lambda2 (IL-28A), and IFN-lambda3 (IL-28B). (22, 23). IFN lambdas are potent antivirals affecting RSV and influenza virus replication, a feature that may also affect influenza reassortment in susceptible cells (24).

ROLE FOR RSV G PROTEIN IN MODULATING HOST RESPONSES

Airway epithelial cells are important in recruiting and activating immune cells in the defense of viral infection. These cells express patterns of Th1- and Th-2 type cytokines and chemokines having a wide range of effects on both innate and adaptive processes. RSV interferes with the host antiviral cytokine response (7). Studies have shown that RSV nonstructural proteins, NS1 and NS2 antagonize the IFN response in infected epithelial cells and affect dendritic cell maturation (25-27). The RSV G protein has also been shown to affect the pattern and magnitude of cytokines and chemokines expressed in the lung following infection of mice (28-30), as well as the trafficking of immune cells (30-32). Recent evidence indicates that RSV G protein governance of the host cell response is associated with let-7 microRNA (miRNA) expression mediated by the central conserved (CX3C) region in the G protein (33). Significantly, RSV infection of a human alveolar epithelial cell line (A549) induced five miRNAs (let-7f, miR-24, miR-337-3p, miR-26b and miR-520a-5p) and repressed two miRNAs (miR-198 and miR-595) whose targets included cell-cycle genes (CCND1, DYRK2 and ELF4), a chemokine gene (CCL7) and SOCS3. Modulating let-7 miRNA levels with miRNA mimics and inhibitors affected RSV replication indicating that RSV modulates host miRNA expression to affect the outcome of the antiviral host response, and this was mediated in part through RSV G protein expression.

Numerous mouse and some human studies have revealed that RSV proteins modulate many aspects of the immune response to infection, particularly the RSV G protein (7). A primary contributor to G protein immune modulation and disease pathogenesis is CX3C chemokine mimicry mediated by the CX3C motif in the central conserved region of the G protein (34). The G protein binds to the fractalkine receptor, CX3CR1, and G protein CX3C-CX3CR1 interaction facilitates virus infection, modulates leukocyte chemotaxis, and adversely affects RSV-specific cytotoxic T cell responses, and enhances disease pathogenesis (31, 34-37). Several studies have shown that antibodies that block G protein CX3C-CX3CR1 interaction protect against G protein-associated pulmonary inflammation, disease outcome, and that they are neutralizing and protective against RSV challenge (38-42). The importance of having anti-RSV G protein antibody responses were revealed in a human study showing that anti-RSV G protein antibody responses after recent RSV infection or vaccination are associated with inhibition of RSV G protein CX3C-CX3CR1 interaction and RSV G protein-mediated leukocyte chemotaxis (43). These findings suggest a vaccination strategy to target antibodies against the G protein could be efficacious. Indeed, it was shown that G protein vaccination to induce antibodies blocking the G protein CX3C-CX3CR1 interaction reduces pulmonary inflammation and virus replication in mice (38), and that mice immunized with RSV A2 G polypeptides generate antibodies that block binding of RSV A2 and B1 native G proteins to CX3CR1, and that these antibodies effectively cross-neutralize both A and B strains of RSV (44).

DEVELOPMENT OF DISEASE INTERVENTION STRATEGIES

Current disease intervention strategies for RSV are limited and largely ineffective, a feature true for influenza as well (45). Protection via vaccination has been unsuccessful for RSV, and is complicated for influenza by the rapid evolution linked to mutilation (antigenic drift) and reassortment (antigenic shift), features that mediate drug resistance (46). Thus, new disease intervention strategies are urgently required, and an important first step is to increase our understanding of the virus-host interface. To do this, studies have been performed to harness the power of RNA interference (RNAi) through the use of small interfering RNA (siRNA) to either target the virus genes directly for silencing (47-49), or the host genes required for virus replication (50-53). Controlling RNAi can be used to provide a powerful platform-enabling technology that will allow for the identification and validation of virus-host interactions and pathways (50, 51, 54, 55). The pro- and anti-viral genes discovered by RNAi screening (53, 56, 57), provides an opportunity to transition the findings toward translational disease intervention approaches such as creation of novel therapeutics, drug repositioning, or create a new generation of vaccine cell lines capable of increased viral production.

FUTURE PERSPECTIVES

The airway epithelium has a central role in defense against respiratory virus infections by expression of antiviral and immune signals mechanisms that contribute to viral resistance and clearance. However, respiratory viruses have evolved strategies to subvert the defense mechanisms leading to cytopathogenesis, increased inflammatory responses, and disease. A thorough understanding of the mechanisms by which viruses modify innate and adaptive immune responses is needed to develop new therapeutic approaches to treat or prevent respiratory diseases caused by important human viral infections mediated by RSV and influenza viruses.

Notes

The author has no financial conflict of interest.

Abbreviations

Ad adenovirus
flu influenza virus
hCoV human coronavirus
HMPV human metapneumovirus
MDA5 melanoma differentiation-associated protein 5
PIV parainfluenza virus
PRRs pattern recognition receptors
RIG-I retinoic acid-inducible gene 1
RSV respiratory syncytial virus
RV rhinovirus
SOCS suppressor of cytokine signaling protein

References

    1. Cate TR. Impact of influenza and other community-acquired viruses. Semin Respir Infect 1998;13:17–23.
    1. Greenberg SB. Rhinovirus and coronavirus infections. Semin Respir Crit Care Med 2007;28:182–192.
    1. Heikkinen T. Role of viruses in the pathogenesis of acute otitis media. Pediatr Infect Dis J 2000;19:S17–S22.
    1. Mahony JB, Petrich A, Smieja M. Molecular diagnosis of respiratory virus infections. Crit Rev Clin Lab Sci 2011;48:217–249.
    1. See H, Wark P. Innate immune response to viral infection of the lungs. Paediatr Respir Rev 2008;9:243–250.
    1. Bartlett JA, Fischer AJ, McCray PB Jr. Innate immune functions of the airway epithelium. Contrib Microbiol 2008;15:147–163.
    1. Oshansky CM, Zhang W, Moore E, Tripp RA. The host response and molecular pathogenesis associated with respiratory syncytial virus infection. Future Microbiol 2009;4:279–297.
    1. Schwarze J, Mackenzie KJ. Novel insights into immune and inflammatory responses to respiratory viruses. Thorax 2013;68:108–110.
    1. Sajjan US. Susceptibility to viral infections in chronic obstructive pulmonary disease: role of epithelial cells. Curr Opin Pulm Med 2013;19:125–132.
    1. Averett DR, Fletcher SP, Li W, Webber SE, Appleman JR. The pharmacology of endosomal TLR agonists in viral disease. Biochem Soc Trans 2007;35:1468–1472.
    1. Sandor F, Buc M. Toll-like receptors. II. Distribution and pathways involved in TLR signalling. Folia Biol (Praha) 2005;51:188–197.
    1. Kurt-Jones EA, Popova L, Kwinn L, Haynes LM, Jones LP, Tripp RA, Walsh EE, Freeman MW, Golenbock DT, Anderson LJ, Finberg RW. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat Immunol 2000;1:398–401.
    1. Haynes LM, Moore DD, Kurt-Jones EA, Finberg RW, Anderson LJ, Tripp RA. Involvement of toll-like receptor 4 in innate immunity to respiratory syncytial virus. J Virol 2001;75:10730–10737.
    1. Awomoyi AA, Rallabhandi P, Pollin TI, Lorenz E, Sztein MB, Boukhvalova MS, Hemming VG, Blanco JC, Vogel SN. Association of TLR4 polymorphisms with symptomatic respiratory syncytial virus infection in high-risk infants and young children. J Immunol 2007;179:3171–3177.
    1. Goodbourn S, Randall RE. The regulation of type I interferon production by paramyxoviruses. J Interferon Cytokine Res 2009;29:539–547.
    1. Le Goffic R, Pothlichet J, Vitour D, Fujita T, Meurs E, Chignard M, Si-Tahar M. Cutting Edge: Influenza A virus activates TLR3-dependent inflammatory and RIG-I-dependent antiviral responses in human lung epithelial cells. J Immunol 2007;178:3368–3372.
    1. Dempoya J, Matsumiya T, Imaizumi T, Hayakari R, Xing F, Yoshida H, Okumura K, Satoh K. Double-stranded RNA induces biphasic STAT1 phosphorylation by both type I interferon (IFN)-dependent and type I IFN-independent pathways. J Virol 2012;86:12760–12769.
    1. O'Neill LA, Bowie AG. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat Rev Immunol 2007;7:353–364.
    1. Oshansky CM, Krunkosky TM, Barber J, Jones LP, Tripp RA. Respiratory syncytial virus proteins modulate suppressors of cytokine signaling 1 and 3 and the type I interferon response to infection by a toll-like receptor pathway. Viral Immunol 2009;22:147–161.
    1. Moore EC, Barber J, Tripp RA. Respiratory syncytial virus (RSV) attachment and nonstructural proteins modify the type I interferon response associated with suppressor of cytokine signaling (SOCS) proteins and IFN-stimulated gene-15 (ISG15). Virol J 2008;5:116.
    1. Tripp RA, Oshansky C, Alvarez R. Cytokines and respiratory syncytial virus infection. Proc Am Thorac Soc 2005;2:147–149.
    1. Lopusna K, Rezuchova I, Betakova T, Skovranova L, Tomaskova J, Lukacikova L, Kabat P. Interferons lambda, new cytokines with antiviral activity. Acta Virol 2013;57:171–179.
    1. Kotenko SV. IFN-lambdas. Curr Opin Immunol 2011;23:583–590.
    1. Hauser MJ, Dlugolenski D, Culhane MR, Wentworth DE, Tompkins SM, Tripp RA. Antiviral responses by Swine primary bronchoepithelial cells are limited compared to human bronchoepithelial cells following influenza virus infection. PLoS One 2013;8:e70251.
    1. Teng MN. The non-structural proteins of RSV: targeting interferon antagonists for vaccine development. Infect Disord Drug Targets 2012;12:129–137.
    1. Thornburg NJ, Hayward SL, Crowe JE Jr. Respiratory syncytial virus regulates human microRNAs by using mechanisms involving beta interferon and NF-kappaB. MBio 2012;3:e00220–e00212.
    1. Munir S, Le Nouen C, Luongo C, Buchholz UJ, Collins PL, Bukreyev A. Nonstructural proteins 1 and 2 of respiratory syncytial virus suppress maturation of human dendritic cells. J Virol 2008;82:8780–8796.
    1. Tripp RA, Jones L, Anderson LJ. Respiratory syncytial virus G and/or SH glycoproteins modify CC and CXC chemokine mRNA expression in the BALB/c mouse. J Virol 2000;74:6227–6229.
    1. Tripp RA, Moore D, Anderson LJ. TH(1)- and TH(2)-TYPE cytokine expression by activated t lymphocytes from the lung and spleen during the inflammatory response to respiratory syncytial virus. Cytokine 2000;12:801–807.
    1. Tripp RA, Moore D, Jones L, Sullender W, Winter J, Anderson LJ. Respiratory syncytial virus G and/or SH protein alters Th1 cytokines, natural killer cells, and neutrophils responding to pulmonary infection in BALB/c mice. J Virol 1999;73:7099–7107.
    1. Harcourt J, Alvarez R, Jones LP, Henderson C, Anderson LJ, Tripp RA. Respiratory syncytial virus G protein and G protein CX3C motif adversely affect CX3CR1+ T cell responses. J Immunol 2006;176:1600–1608.
    1. Tripp RA, Anderson LJ. Cytotoxic T-lymphocyte precursor frequencies in BALB/c mice after acute respiratory syncytial virus (RSV) infection or immunization with a formalin-inactivated RSV vaccine. J Virol 1998;72:8971–8975.
    1. Bakre A, Mitchell P, Coleman JK, Jones LP, Saavedra G, Teng M, Tompkins SM, Tripp RA. Respiratory syncytial virus modifies microRNAs regulating host genes that affect virus replication. J Gen Virol 2012;93:2346–2356.
    1. Tripp RA, Jones LP, Haynes LM, Zheng H, Murphy PM, Anderson LJ. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nat Immunol 2001;2:732–738.
    1. Haynes LM, Jones LP, Barskey A, Anderson LJ, Tripp RA. Enhanced disease and pulmonary eosinophilia associated with formalin-inactivated respiratory syncytial virus vaccination are linked to G glycoprotein CX3C-CX3CR1 interaction and expression of substance P. J Virol 2003;77:9831–9844.
    1. Tripp RA, Dakhama A, Jones LP, Barskey A, Gelfand EW, Anderson LJ. The G glycoprotein of respiratory syncytial virus depresses respiratory rates through the CX3C motif and substance P. J Virol 2003;77:6580–6584.
    1. Li XQ, Fu ZF, Alvarez R, Henderson C, Tripp RA. Respiratory syncytial virus (RSV) infects neuronal cells and processes that innervate the lung by a process involving RSV G protein. J Virol 2006;80:537–540.
    1. Zhang W, Choi Y, Haynes LM, Harcourt JL, Anderson LJ, Jones LP, Tripp RA. Vaccination to induce antibodies blocking the CX3C-CX3CR1 interaction of respiratory syncytial virus G protein reduces pulmonary inflammation and virus replication in mice. J Virol 2010;84:1148–1157.
    1. Kauvar LM, Harcourt JL, Haynes LM, Tripp RA. Therapeutic targeting of respiratory syncytial virus G-protein. Immunotherapy 2010;2:655–661.
    1. Miao C, Radu GU, Caidi H, Tripp RA, Anderson LJ, Haynes LM. Treatment with respiratory syncytial virus G glycoprotein monoclonal antibody or F(ab')2 components mediates reduced pulmonary inflammation in mice. J Gen Virol 2009;90:1119–1123.
    1. Haynes LM, Caidi H, Radu GU, Miao C, Harcourt JL, Tripp RA, Anderson LJ. Therapeutic monoclonal antibody treatment targeting respiratory syncytial virus (RSV) G protein mediates viral clearance and reduces the pathogenesis of RSV infection in BALB/c mice. J Infect Dis 2009;200:439–447.
    1. Collarini EJ, Lee FE, Foord O, Park M, Sperinde G, Wu H, Harriman WD, Carroll SF, Ellsworth SL, Anderson LJ, Tripp RA, Walsh EE, Keyt BA, Kauvar LM. Potent high-affinity antibodies for treatment and prophylaxis of respiratory syncytial virus derived from B cells of infected patients. J Immunol 2009;183:6338–6345.
    1. Harcourt JL, Karron RA, Tripp RA. Anti-G protein antibody responses to respiratory syncytial virus infection or vaccination are associated with inhibition of G protein CX3C-CX3CR1 binding and leukocyte chemotaxis. J Infect Dis 2004;190:1936–1940.
    1. Choi Y, Mason CS, Jones LP, Crabtree J, Jorquera PA, Tripp RA. Antibodies to the central conserved region of respiratory syncytial virus (RSV) G protein block RSV G protein CX3C-CX3CR1 binding and cross-neutralize RSV A and B strains. Viral Immunol 2012;25:193–203.
    1. Beeler JA, Eichelberger MC. Influenza and respiratory syncytial virus (RSV) vaccines for infants: safety, immunogenicity, and efficacy. Microb Pathog 2013;55:9–15.
    1. Schmidt AC. Progress in respiratory virus vaccine development. Semin Respir Crit Care Med 2011;32:527–540.
    1. DeVincenzo JP. The promise, pitfalls and progress of RNA-interference-based antiviral therapy for respiratory viruses. Antivir Ther 2012;17:213–225.
    1. Alvarez R, Elbashir S, Borland T, Toudjarska I, Hadwiger P, John M, Roehl I, Morskaya SS, Martinello R, Kahn J, Van Ranst M, Tripp RA, DeVincenzo JP, Pandey R, Maier M, Nechev L, Manoharan M, Kotelianski V, Meyers R. RNA interference-mediated silencing of the respiratory syncytial virus nucleocapsid defines a potent antiviral strategy. Antimicrob Agents Chemother 2009;53:3952–3962.
    1. Zhang W, Tripp RA. RNA interference inhibits respiratory syncytial virus replication and disease pathogenesis without inhibiting priming of the memory immune response. J Virol 2008;82:12221–12231.
    1. Bakre A, Andersen LE, Meliopoulos V, Coleman K, Yan X, Brooks P, Crabtree J, Tompkins SM, Tripp RA. Identification of host kinase genes required for influenza virus replication and the regulatory role of MicroRNAs. PLoS One 2013;8:e66796.
    1. Perwitasari O, Yan X, Johnson S, White C, Brooks P, Tompkins SM, Tripp RA. Targeting organic anion transporter 3 with probenecid as a novel anti-influenza a virus strategy. Antimicrob Agents Chemother 2013;57:475–483.
    1. Meliopoulos VA, Andersen LE, Brooks P, Yan X, Bakre A, Coleman JK, Tompkins SM, Tripp RA. MicroRNA regulation of human protease genes essential for influenza virus replication. PLoS One 2012;7:e37169.
    1. Meliopoulos VA, Andersen LE, Birrer KF, Simpson KJ, Lowenthal JW, Bean AG, Stambas J, Stewart CR, Tompkins SM, van Beusechem VW, Fraser I, Mhlanga M, Barichievy S, Smith Q, Leake D, Karpilow J, Buck A, Jona G, Tripp RA. Host gene targets for novel influenza therapies elucidated by high-throughput RNA interference screens. FASEB J 2012;26:1372–1386.
    1. Tran AT, Rahim MN, Ranadheera C, Kroeker A, Cortens JP, Opanubi KJ, Wilkins JA, Coombs KM. Knockdown of specific host factors protects against influenza virus-induced cell death. Cell Death Dis 2013;4:e769.
    1. Prusty BK, Karlas A, Meyer TF, Rudel T. Genome-wide RNAi screen for viral replication in mammalian cell culture. Methods Mol Biol 2011;721:383–395.
    1. Panda D, Cherry S. Cell-based genomic screening: elucidating virus-host interactions. Curr Opin Virol 2012;2:784–792.
    1. Kassner PD. Discovery of novel targets with high throughput RNA interference screening. Comb Chem High Throughput Screen 2008;11:175–184.

Metrics
Share
PERMALINK