Next Article in Journal
Interplay between the Lung Microbiome, Pulmonary Immunity and Viral Reservoirs in People Living with HIV under Antiretroviral Therapy
Next Article in Special Issue
Enhancing Anti-G Antibody Induction by a Live Single-Cycle Prefusion F—Expressing RSV Vaccine Improves In Vitro and In Vivo Efficacy
Previous Article in Journal
Interspecies Transmission from Pigs to Ferrets of Antigenically Distinct Swine H1 Influenza A Viruses with Reduced Reactivity to Candidate Vaccine Virus Antisera as Measures of Relative Zoonotic Risk
Previous Article in Special Issue
Detailed Molecular Interactions between Respiratory Syncytial Virus Fusion Protein and the TLR4/MD-2 Complex In Silico
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

RSV Replication, Transmission, and Disease Are Influenced by the RSV G Protein

by
Harrison C. Bergeron
and
Ralph A. Tripp
*
Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30605, USA
*
Author to whom correspondence should be addressed.
Viruses 2022, 14(11), 2396; https://doi.org/10.3390/v14112396
Submission received: 26 September 2022 / Revised: 24 October 2022 / Accepted: 28 October 2022 / Published: 29 October 2022
(This article belongs to the Special Issue Respiratory Syncytial Virus 2.0)

Abstract

:
It is important to understand the features affecting virus replication, fitness, and transmissibility as they contribute to the outcome of infection and affect disease intervention approaches. Respiratory syncytial virus (RSV) is a major contributor to respiratory disease, particularly in the infant and elderly populations. Although first described over 60 years ago, there are no approved vaccines and there are limited specific antiviral treatments due in part to our incomplete understanding of the features affecting RSV replication, immunity, and disease. RSV studies have typically focused on using continuous cell lines and conventional RSV strains to establish vaccine development and various antiviral countermeasures. This review outlines how the RSV G protein influences viral features, including replication, transmission, and disease, and how understanding the role of the G protein can improve the understanding of preclinical studies.

1. Overview of RSV

Understanding the features that contribute to RSV replication, fitness, and transmissibility is important for understanding the outcomes of infection and disease intervention approaches. RSV infects young children, with many experiencing more than one infection by 2 years of age [1,2]. Globally, RSV causes acute respiratory infection in infants and young children, leading to >60,000 in-hospital deaths and >3 million hospital admissions per year in children <5 years old [3]. The RSV disease burden in high-risk adults with chronic medical conditions is high and similar to non-pandemic influenza [4]. The RSV genome is a negative-sense single-stranded RNA, is 15.2 kb in length, and contains 10 genes encoding 11 proteins [5]. The RSV G and F proteins are the two major surface proteins and have key roles in virus entry, replication, and immune modulation. The attachment G protein binds to ciliated respiratory epithelial cells, and the F protein mediates viral fusion with the target cell membranes [6,7]. The F protein is a major target for antiviral drug and vaccine development, and both the G and F proteins are the antigens targeted by neutralizing antibodies induced by infection. The G protein is produced either as a membrane-bound or secreted protein (e.g., soluble G) that mediates immune evasion [8,9]. The F protein is a trimer in the infected cell membrane in its prefusion form but refolds into a post-fusion form. The SH protein is a minor surface protein in RSV and is expressed at low levels [10]. RSV F, G, and SH proteins can form an oligomeric complex within infected cells, and depending on the host cell, exist as oligomeric forms on the surface of RSV [11,12,13]. RSV is divided into two antigenic groups, i.e., A and B, according to the epitope differences mainly in the G protein [14]. The G protein is the most variable protein [15]. Despite differences in the G protein, it contains a central conserved domain (CCD) and is a CX3C chemokine motif that lacks glycosylation and functions as a chemokine mimic [16,17].
RNA viruses such as RSV replicate using low-fidelity polymerases leading to mutation rates that result in a virus cloud with a distribution of mutants [18]. The spontaneous mutations may alter virus replication leading to the emergence of variant viruses with a selective advantage, i.e., increased fitness [19]. RSV variants can also arise due to selective pressures often by the growth of the virus in the presence of antibodies. These fitness differences in RSV have been linked to nucleotide substitutions in the F gene mediated by anti-F protein antibodies and perhaps mutations occurring elsewhere in the genome [20,21]. Substantial efforts to understand the mechanisms by which virus fitness is increased or decreased are important in vaccine design [14]. Fitness outcomes are measured by both biological (plaque assay) and molecular techniques (nucleotide sequence determination). To understand RSV fitness, one must consider the important steps required in replicating the virus, i.e., binding host cell receptors, entry, virus transcription, translation, and genome replication. The cycle of RSV replication begins with G protein-mediated attachment to the apical surface of polarized, ciliated airway epithelial cells [15], and is facilitated by the G protein CX3C binding to CX3CR1, as well as potentially other cell surface molecules [22,23]. Viral entry is enabled by F protein-mediated membrane fusion [24]. Transcription and replication occur in the host cell cytoplasm in viral inclusion bodies [25]. The viral RNA is transcribed by the viral RNA-dependent RNA polymerase complex and into the positive-sense anti-genome intermediates needed for the replication of new negative-sense genomes for virus packaging [26], where the assembly of RSV occurs at or near the plasma membrane [27].

2. RSV Infection Is Cell-Type Dependent

RSV infection occurs following virus inhalation, leading to small foci of infection on the apical surface of respiratory airway cells [28]. This process stimulates the pattern recognition receptors (PRRs), primarily retinoic acid-inducible gene I (RIG-I), but also several Toll-like receptors including TLR2 and TLR4 [29,30,31,32,33]. The RSV F protein has been shown to activate cells through TLR4 [30], whereas the RSV G protein modifies and reduces TLR activation [29,34], and infants with TLR4 SNPs have been shown to be at greater risk for severe RSV disease [35]. TLR and PRR activation initiates host innate immune responses that include expression of type I and III IFNs, IFN-stimulated genes (ISGs), and a variety of proinflammatory cytokines and chemokines [36,37]. To gain a better understanding of RSV and the host response to infection, most studies have typically examined RSV strain A infection of continuous or transformed cell lines such as Hep-2 cells, A549 cells, and Vero cells; however, the results often do not foretell the outcomes from primary human epithelial cell systems or with clinical RSV isolates, particularly as there are several RSV proteins that modify nonspecific (e.g., interferons) and specific (e.g., antibodies) factors affecting virus replication.
Hep-2 (human epithelial) cells were originally derived from a human larynx carcinoma and have shown evidence of contamination with HeLa cells [38]. A549 cells are type II lung carcinoma epithelial cells and were derived from an epithelial carcinoma [39]. One study showed important RSV subgroup-dependent differences in the viral gene expression between RSV-infected Hep-2 and A549 cells [40]. Specifically, Hep-2 cells had increased viral titers and host gene expression compared to RSV-infected A549 cells, suggesting that the origin of the host cell has a dominant effect, whereas the infecting RSV strain has a smaller role in the outcome of an RSV infection. These data revealed important differences in the host response to RSV infection between two widely used continuous cell lines, suggesting the need to evaluate relevant human cells and clinical viruses. Vero cells, derived from the kidney of an African green monkey, are one of the more commonly used continuous cell lines [41]. These cell lines have known differences in RSV propagation outcomes in which the altered expression of viral proteins can occur, as well as differences in glycosylation [12,40]. Most changes have been associated with the cell-specific glycosylation differences of the G protein, which have been demonstrated using lectins and carbohydrate-specific antibodies [11]. For example, when RSV is propagated in Vero cells, the predicted molecular mass of the G protein is ~55 kDa [12], and in Hep-2 cells, the G protein is ~90 kDa, but when RSV is propagated in primary cultures, the G protein is ~170 kDa [42]. In addition, it is important to be cognizant that RSV grown in Vero cells may contain a truncated attachment protein that alters RSV infectivity and dependence on glycosaminoglycans, including heparan sulfate (HS) [12]. This dependence on glycosaminoglycans for initiating infection does not recapitulate wild-type RSV infection, as HS is absent in human airway epithelial cells and infection occurs mainly via specific receptor binding [43]. These G protein modifications affect RSV infection, permissibility, and the host response to infection. Thus, it is important to consider the cell type used for RSV propagation, as continuous cells are not necessarily interchangeable with respect to their permissiveness, their sensitivity to different RSV strains, or the host cell response to infection.
Differences in RSV susceptibility and replication exist between continuous cell lines and normal human cells, particularly between infection with laboratory isolates compared to clinical RSV isolates. A useful immortalized but non-tumorigenic human respiratory epithelial cell line is the BEAS-2B cell line, which has been used to compare RSV infection and host cell responses [44]. BEAS-2B cells are highly permissive to RSV infection but have restricted RSV replication, which has been linked to their antiviral response that is associated with type I and III IFN expression [45]. RSV-infected BEAS-2B cells respond similarly to RSV-infected primary human respiratory epithelial cells in which the differential expression of type I and III IFNs, ISGs, and proinflammatory cytokines is expressed [46]. The most susceptible to RSV infection are Vero cells, which lack the expression of type I IFNs while retaining type I receptors [47]. Differences in RSV susceptibility are not exclusive to cell lines. Genetic differences in mice have also revealed strain-dependent differences. For example, RSV titers are considerably higher in AKR/J mice, which are permissive compared with C57BL/6J mice, which are resistant [48]. Some inbred mice, e.g., BALB/c mice, have silenced Mx-1 genes, making them more susceptible to viral infections [49]. The interferon-inducible Mx protein is responsible for a specific antiviral state against virus infection [50]. Thus, it is important to consider genetic differences and how this may affect the results.

3. RSV Fitness Is Strain Dependent

The findings that RSV is altered by the cell line or cell type during infection show that the host cell can modify viral fitness and permissibility that can be dependent on the infecting strain. The strain of RSV is also important to consider. For example, RSV strain A2 was first isolated in 1961 from the lower respiratory tract of an infant [51] and is the predominant strain used for the development of RSV vaccine candidates. There have been limited in vitro or immunology studies examined for RSV strain B (RSV/B) despite a similar circulation to strain A [51,52,53,54]. RSV strain differences are a determinant of disease phenotypes and severity [55]; thus, it is important to study strain differences in response to infection. RSV line 19 is an RSV A strain that was first isolated from an infant with respiratory illness [56]. RSV A strains (A2, Long) have been widely used laboratory strains used in small animal studies of RSV pathogenesis but inadequately emulate RSV disease. In contrast, the RSV clinical strain, i.e., Line19, induces airway mucus expression in BALB/c mice, as well as lung dysfunction and mucin expression [57], and the chimeric Line19F has improved thermostability and further induces a mucogenic response in mice [58]. Another RSV clinical strain commonly used for clinical trials is the Memphis-37 strain, which is an A strain isolated from a child with bronchiolitis [59]. Memphis-37 has been used to study RSV pathogenesis and test vaccines and RSV inhibitors in human challenge studies [51,60,61]. Although uncommonly used in pre-clinical studies or clinical trials, the two circulating RSV strains ON (RSV A) and BA (RSV B) contain duplications in the G protein, which likely improve viral fitness [60,62]. It is evident that an over-reliance on one RSV strain, which inadequately recapitulates RSV disease in mice (e.g., A2), may overstate or understate drug or vaccine efficacy.

4. G protein Functions as the Attachment and Immune Modifier Protein

Many RSV proteins can modify the host cell response to infection [37]. RSV interacts with the host to create a favorable environment for virus replication and transmission ultimately making the host more susceptible to infection. For example, the G protein has attributes that contribute to mimicry and immune evasion [63,64,65,66,67]. The soluble form of the G protein differs from the membrane-bound form in its oligomeric state but remains capable of binding to cell surface glycosaminoglycans [68]. An excess of the G protein is expressed as a soluble G in a form that functions as an antigenic decoy, and this molecule retains the same characteristics as Gm based on glycosylation and antibody reactivity [69]. The membrane-bound G protein is a type II glycosylated membrane protein that has an extracellular ectodomain containing a central conserved domain (CCD) with four cysteine residues that are highly conserved in all RSV isolates. The CCD contains a CX3C chemokine motif (amino acids 182–186) that facilitates RSV attachment to susceptible cells bearing a CX3C chemokine receptor, CX3CR1 [22]. CX3CR1 mimicry by the G protein facilitates RSV infection and may alter CX3CL1 (fractalkine) chemotaxis of human and mouse leukocytes [22,70]. Expression of the G protein during RSV infection of mice has also been shown to decrease the number of activated and RSV-specific pulmonary CX3CR1+ T cells, as well as natural killer (NK) cells [70,71]. These studies suggest that the RSV G protein can modulate immune responses. Additionally, there is a link between the RSV G protein, other RSV proteins, and IFN expression, particularly for NS proteins [72,73,74]. As type I IFNs typically sensitize infected cells to programmed cell death, type III IFNs induce an anti-viral state. RSV infection induces high expression levels of IFNλ, which in one recently study was shown as protective [75], while another associated type III IFNs with more severe disease in children [76]. Thus, the RSV G protein may affect virus replication, transmission, immune responses, and disease.

5. Conclusions

Despite the several decades since RSV was first identified, there remains no safe and approved vaccine [77]. Prophylaxis is restricted to high-risk infants and has modest efficacy, and there is a lack of post-exposure therapeutic options. The chief reason for this lag is our incomplete understanding of RSV infection and disease. Current advances have led to the development of several promising vaccines and drug candidates. However, it is likely that more than one type of RSV vaccine will be needed to immunize those who could benefit from vaccination. In addition, more translational RSV studies need to be performed using primary or normal human cell lines and types infected with clinically relevant RSV strains to better understand the features contributing to replication, infectivity, transmission, and disease pathogenesis. It will also be important to consider vaccines and treatments that target both the RSV F and G proteins, as several preclinical studies have shown that anti-G protein antibodies both neutralize RSV infection in vivo and modify RSV disease [63,78,79,80,81,82,83,84], as well as human airway epithelial cells.

Author Contributions

Both authors contributed equally to the writing and editing of this review. All authors have read and agreed to the published version of the manuscript.

Funding

Funding was provided by Tripp Lab and the Georgia Research Alliance (GRA).

Conflicts of Interest

Authors have no conflicts of interest to declare.

References

  1. Glezen, W.P.; Taber, L.H.; Frank, A.L.; Kasel, J.A. Risk of primary infection and reinfection with respiratory syncytial virus. Am. J. Dis. Child. 1986, 140, 543–546. [Google Scholar] [CrossRef] [PubMed]
  2. Zylbersztejn, A.; Pembrey, L.; Goldstein, H.; Berbers, G.; Schepp, R.; van der Klis, F.; Sande, C.; Mason, D.; Wright, J.; Smyth, R.; et al. Respiratory syncytial virus in young children: Community cohort study integrating serological surveys, questionnaire and electronic health records, Born in Bradford cohort, England, 2008 to 2013. Eurosurveillance 2021, 26, 2000023. [Google Scholar] [CrossRef] [PubMed]
  3. Andeweg, S.P.; Schepp, R.M.; van de Kassteele, J.; Mollema, L.; Berbers, G.A.M.; van Boven, M. Population-based serology reveals risk factors for RSV infection in children younger than 5 years. Sci. Rep. 2021, 11, 8953. [Google Scholar] [CrossRef] [PubMed]
  4. Falsey, A.R.; Hennessey, P.A.; Formica, M.A.; Cox, C.; Walsh, E.E. Respiratory syncytial virus infection in elderly and high-risk adults. N. Engl. J. Med. 2005, 352, 1749–1759. [Google Scholar] [CrossRef] [PubMed]
  5. Battles, M.B.; McLellan, J.S. Respiratory syncytial virus entry and how to block it. Nat. Rev. Microbiol. 2019, 17, 233–245. [Google Scholar] [CrossRef]
  6. Levine, S.; Klaiber-Franco, R.; Paradiso, P.R. Demonstration that glycoprotein G is the attachment protein of respiratory syncytial virus. J. Gen. Virol. 1987, 68, 2521–2524. [Google Scholar] [CrossRef]
  7. McLellan, J.S.; Ray, W.C.; Peeples, M.E. Structure and function of respiratory syncytial virus surface glycoproteins. Curr. Top. Microbiol. Immunol. 2013, 372, 83–104. [Google Scholar] [CrossRef] [Green Version]
  8. Efstathiou, C.; Abidi, S.H.; Harker, J.; Stevenson, N.J. Revisiting respiratory syncytial virus’s interaction with host immunity, towards novel therapeutics. Cell. Mol. Life Sci. 2020, 77, 5045–5058. [Google Scholar] [CrossRef]
  9. Ouyang, Y.; Liao, H.; Hu, Y.; Luo, K.; Hu, S.; Zhu, H. Innate Immune Evasion by Human Respiratory Syncytial Virus. Front. Microbiol. 2022, 13, 865592. [Google Scholar] [CrossRef]
  10. Rixon, H.W.M.; Brown, G.; Aitken, J.; McDonald, T.; Graham, S.; Sugrue, R.J. The small hydrophobic (SH) protein accumulates within lipid-raft structures of the Golgi complex during respiratory syncytial virus infection. J. Gen. Virol. 2004, 85, 1153–1165. [Google Scholar] [CrossRef]
  11. Garcia-Beato, R.; Martinez, I.; Franci, C.; Real, F.X.; Garcia-Barreno, B.; Melero, J.A. Host cell effect upon glycosylation and antigenicity of human respiratory syncytial virus G glycoprotein. Virology 1996, 221, 301–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kwilas, S.; Liesman, R.M.; Zhang, L.; Walsh, E.; Pickles, R.J.; Peeples, M.E. Respiratory syncytial virus grown in Vero cells contains a truncated attachment protein that alters its infectivity and dependence on glycosaminoglycans. J. Virol. 2009, 83, 10710–10718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Feldman, S.A.; Crim, R.L.; Audet, S.A.; Beeler, J.A. Human respiratory syncytial virus surface glycoproteins F, G and SH form an oligomeric complex. Arch. Virol. 2001, 146, 2369–2383. [Google Scholar] [CrossRef]
  14. Yu, J.M.; Fu, Y.H.; Peng, X.L.; Zheng, Y.P.; He, J.S. Genetic diversity and molecular evolution of human respiratory syncytial virus A and B. Sci. Rep. 2021, 11, 12941. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, L.; Peeples, M.E.; Boucher, R.C.; Collins, P.L.; Pickles, R.J. Respiratory syncytial virus infection of human airway epithelial cells is polarized, specific to ciliated cells, and without obvious cytopathology. J. Virol. 2002, 76, 5654–5666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kauvar, L.M.; Harcourt, J.L.; Haynes, L.M.; Tripp, R.A. Therapeutic targeting of respiratory syncytial virus G-protein. Immunotherapy 2010, 2, 655–661. [Google Scholar] [CrossRef] [Green Version]
  17. Jorquera, P.A.; Anderson, L.; Tripp, R.A. Understanding respiratory syncytial virus (RSV) vaccine development and aspects of disease pathogenesis. Expert Rev. Vaccines 2016, 15, 173–187. [Google Scholar] [CrossRef]
  18. Mandary, M.B.; Masomian, M.; Poh, C.L. Impact of RNA Virus Evolution on Quasispecies Formation and Virulence. Int. J. Mol. Sci. 2019, 20, 4657. [Google Scholar] [CrossRef] [Green Version]
  19. Steinhauer, D.A.; Holland, J.J. Rapid evolution of RNA viruses. Annu. Rev. Microbiol. 1987, 41, 409–433. [Google Scholar] [CrossRef]
  20. Zhao, X.; Liu, E.; Chen, F.P.; Sullender, W.M. In vitro and in vivo fitness of respiratory syncytial virus monoclonal antibody escape mutants. J. Virol. 2006, 80, 11651–11657. [Google Scholar] [CrossRef]
  21. Langedijk, A.C.; Lebbink, R.J.; Naaktgeboren, C.; Evers, A.; Viveen, M.C.; Greenough, A.; Heikkinen, T.; Stein, R.T.; Richmond, P.; Martinon-Torres, F.; et al. Global molecular diversity of RSV—The “INFORM RSV” study. BMC Infect. Dis. 2020, 20, 450. [Google Scholar] [CrossRef] [PubMed]
  22. Tripp, R.A.; Jones, L.P.; Haynes, L.M.; Zheng, H.; Murphy, P.M.; Anderson, L.J. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nat. Immunol. 2001, 2, 732–738. [Google Scholar] [CrossRef] [PubMed]
  23. Johnson, S.M.; McNally, B.A.; Ioannidis, I.; Flano, E.; Teng, M.N.; Oomens, A.G.; Walsh, E.E.; Peeples, M.E. Respiratory Syncytial Virus Uses CX3CR1 as a Receptor on Primary Human Airway Epithelial Cultures. PLoS Pathog. 2015, 11, e1005318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Collins, P.L.; Huang, Y.T.; Wertz, G.W. Nucleotide sequence of the gene encoding the fusion (F) glycoprotein of human respiratory syncytial virus. Proc. Natl. Acad. Sci. USA 1984, 81, 7683–7687. [Google Scholar] [CrossRef] [Green Version]
  25. Garcia, J.; Garcia-Barreno, B.; Vivo, A.; Melero, J.A. Cytoplasmic inclusions of respiratory syncytial virus-infected cells: Formation of inclusion bodies in transfected cells that coexpress the nucleoprotein, the phosphoprotein, and the 22K protein. Virology 1993, 195, 243–247. [Google Scholar] [CrossRef]
  26. Noton, S.L.; Fearns, R. Initiation and regulation of paramyxovirus transcription and replication. Virology 2015, 479–480, 545–554. [Google Scholar] [CrossRef] [Green Version]
  27. Gower, T.L.; Pastey, M.K.; Peeples, M.E.; Collins, P.L.; McCurdy, L.H.; Hart, T.K.; Guth, A.; Johnson, T.R.; Graham, B.S. RhoA signaling is required for respiratory syncytial virus-induced syncytium formation and filamentous virion morphology. J. Virol. 2005, 79, 5326–5336. [Google Scholar] [CrossRef] [Green Version]
  28. Johnson, J.E.; Gonzales, R.A.; Olson, S.J.; Wright, P.F.; Graham, B.S. The histopathology of fatal untreated human respiratory syncytial virus infection. Mod. Pathol. 2007, 20, 108–119. [Google Scholar] [CrossRef] [Green Version]
  29. Murawski, M.R.; Bowen, G.N.; Cerny, A.M.; Anderson, L.J.; Haynes, L.M.; Tripp, R.A.; Kurt-Jones, E.A.; Finberg, R.W. Respiratory syncytial virus activates innate immunity through Toll-like receptor 2. J. Virol. 2009, 83, 1492–1500. [Google Scholar] [CrossRef] [Green Version]
  30. Haynes, L.M.; Moore, D.D.; Kurt-Jones, E.A.; Finberg, R.W.; Anderson, L.J.; Tripp, R.A. Involvement of toll-like receptor 4 in innate immunity to respiratory syncytial virus. J. Virol. 2001, 75, 10730–10737. [Google Scholar] [CrossRef]
  31. Ban, J.; Lee, N.R.; Lee, N.J.; Lee, J.K.; Quan, F.S.; Inn, K.S. Human Respiratory Syncytial Virus NS 1 Targets TRIM25 to Suppress RIG-I Ubiquitination and Subsequent RIG-I-Mediated Antiviral Signaling. Viruses 2018, 10, 716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kurt-Jones, E.A.; Popova, L.; Kwinn, L.; Haynes, L.M.; Jones, L.P.; Tripp, R.A.; Walsh, E.E.; Freeman, M.W.; Golenbock, D.T.; Anderson, L.J.; et al. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat. Immunol. 2000, 1, 398–401. [Google Scholar] [CrossRef] [PubMed]
  33. Alshaghdali, K.; Saeed, M.; Kamal, M.A.; Saeed, A. Interaction of Ectodomain of Respiratory Syncytial Virus G Protein with TLR2/ TLR6 Heterodimer: An In Vitro and In Silico Approach to Decipher the Role of RSV G Protein in Pro-Inflammatory Response against the Virus. Curr. Pharm. Des. 2021, 27, 4464–4476. [Google Scholar] [CrossRef] [PubMed]
  34. Shingai, M.; Azuma, M.; Ebihara, T.; Sasai, M.; Funami, K.; Ayata, M.; Ogura, H.; Tsutsumi, H.; Matsumoto, M.; Seya, T. Soluble G protein of respiratory syncytial virus inhibits Toll-like receptor 3/4-mediated IFN-beta induction. Int. Immunol. 2008, 20, 1169–1180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Awomoyi, A.A.; Rallabhandi, P.; Pollin, T.I.; Lorenz, E.; Sztein, M.B.; Boukhvalova, M.S.; Hemming, V.G.; Blanco, J.C.; Vogel, S.N. Association of TLR4 polymorphisms with symptomatic respiratory syncytial virus infection in high-risk infants and young children. J. Immunol. 2007, 179, 3171–3177. [Google Scholar] [CrossRef] [Green Version]
  36. Stambas, J.; Lu, C.; Tripp, R.A. Innate and adaptive immune responses in respiratory virus infection: Implications for the clinic. Expert Rev. Respir. Med. 2020, 14, 1141–1147. [Google Scholar] [CrossRef]
  37. Oshansky, C.M.; Zhang, W.; Moore, E.; Tripp, R.A. The host response and molecular pathogenesis associated with respiratory syncytial virus infection. Future Microbiol. 2009, 4, 279–297. [Google Scholar] [CrossRef] [Green Version]
  38. Gartler, S.M. Apparent Hela cell contamination of human heteroploid cell lines. Nature 1968, 217, 750–751. [Google Scholar] [CrossRef]
  39. Foster, K.A.; Oster, C.G.; Mayer, M.M.; Avery, M.L.; Audus, K.L. Characterization of the A549 cell line as a type II pulmonary epithelial cell model for drug metabolism. Exp. Cell. Res. 1998, 243, 359–366. [Google Scholar] [CrossRef]
  40. Rajan, A.; Piedra, F.A.; Aideyan, L.; McBride, T.; Robertson, M.; Johnson, H.L.; Aloisio, G.M.; Henke, D.; Coarfa, C.; Stossi, F.; et al. Multiple Respiratory Syncytial Virus (RSV) Strains Infecting HEp-2 and A549 Cells Reveal Cell Line-Dependent Differences in Resistance to RSV Infection. J. Virol. 2022, 96, e0190421. [Google Scholar] [CrossRef]
  41. Ammerman, N.C.; Beier-Sexton, M.; Azad, A.F. Growth and maintenance of Vero cell lines. Curr. Protoc. Microbiol. 2008, 11, A-4E. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. King, T.; Mejias, A.; Ramilo, O.; Peeples, M.E. The larger attachment glycoprotein of respiratory syncytial virus produced in primary human bronchial epithelial cultures reduces infectivity for cell lines. PLoS Pathog. 2021, 17, e1009469. [Google Scholar] [CrossRef] [PubMed]
  43. Zhang, L.; Bukreyev, A.; Thompson, C.I.; Watson, B.; Peeples, M.E.; Collins, P.L.; Pickles, R.J. Infection of ciliated cells by human parainfluenza virus type 3 in an in vitro model of human airway epithelium. J. Virol. 2005, 79, 1113–1124. [Google Scholar] [CrossRef] [Green Version]
  44. Han, X.; Na, T.; Wu, T.; Yuan, B.Z. Human lung epithelial BEAS-2B cells exhibit characteristics of mesenchymal stem cells. PLoS ONE 2020, 15, e0227174. [Google Scholar] [CrossRef]
  45. Hillyer, P.; Shepard, R.; Uehling, M.; Krenz, M.; Sheikh, F.; Thayer, K.R.; Huang, L.; Yan, L.; Panda, D.; Luongo, C.; et al. Differential Responses by Human Respiratory Epithelial Cell Lines to Respiratory Syncytial Virus Reflect Distinct Patterns of Infection Control. J. Virol. 2018, 92, e02202-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Oshansky, C.M.; Barber, J.P.; Crabtree, J.; Tripp, R.A. Respiratory syncytial virus F and G proteins induce interleukin 1alpha, CC, and CXC chemokine responses by normal human bronchoepithelial cells. J. Infect. Dis. 2010, 201, 1201–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Mosca, J.D.; Pitha, P.M. Transcriptional and posttranscriptional regulation of exogenous human beta interferon gene in simian cells defective in interferon synthesis. Mol. Cell. Biol. 1986, 6, 2279–2283. [Google Scholar] [CrossRef]
  48. Stark, J.M.; McDowell, S.A.; Koenigsknecht, V.; Prows, D.R.; Leikauf, J.E.; Le Vine, A.M.; Leikauf, G.D. Genetic susceptibility to respiratory syncytial virus infection in inbred mice. J. Med. Virol. 2002, 67, 92–100. [Google Scholar] [CrossRef]
  49. Jin, H.K.; Yamashita, T.; Ochiai, K.; Haller, O.; Watanabe, T. Characterization and expression of the Mx1 gene in wild mouse species. Biochem. Genet. 1998, 36, 311–322. [Google Scholar] [CrossRef]
  50. Verhelst, J.; Hulpiau, P.; Saelens, X. Mx proteins: Antiviral gatekeepers that restrain the uninvited. Microbiol. Mol. Biol. Rev. 2013, 77, 551–566. [Google Scholar] [CrossRef]
  51. Pandya, M.C.; Callahan, S.M.; Savchenko, K.G.; Stobart, C.C. A Contemporary View of Respiratory Syncytial Virus (RSV) Biology and Strain-Specific Differences. Pathogens 2019, 8, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Melero, J.A.; Moore, M.L. Influence of respiratory syncytial virus strain differences on pathogenesis and immunity. Curr. Top. Microbiol. Immunol. 2013, 372, 59–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Langedijk, A.C.; Harding, E.R.; Konya, B.; Vrancken, B.; Lebbink, R.J.; Evers, A.; Willemsen, J.; Lemey, P.; Bont, L.J. A systematic review on global RSV genetic data: Identification of knowledge gaps. Rev. Med. Virol. 2022, 32, e2284. [Google Scholar] [CrossRef]
  54. Papadopoulos, N.G.; Gourgiotis, D.; Javadyan, A.; Bossios, A.; Kallergi, K.; Psarras, S.; Tsolia, M.N.; Kafetzis, D. Does respiratory syncytial virus subtype influences the severity of acute bronchiolitis in hospitalized infants? Respir. Med. 2004, 98, 879–882. [Google Scholar] [CrossRef] [PubMed]
  55. Stokes, K.L.; Chi, M.H.; Sakamoto, K.; Newcomb, D.C.; Currier, M.G.; Huckabee, M.M.; Lee, S.; Goleniewska, K.; Pretto, C.; Williams, J.V.; et al. Differential pathogenesis of respiratory syncytial virus clinical isolates in BALB/c mice. J. Virol. 2011, 85, 5782–5793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Herlocher, M.L.; Ewasyshyn, M.; Sambhara, S.; Gharaee-Kermani, M.; Cho, D.; Lai, J.; Klein, M.; Maassab, H.F. Immunological properties of plaque purified strains of live attenuated respiratory syncytial virus (RSV) for human vaccine. Vaccine 1999, 17, 172–181. [Google Scholar] [CrossRef]
  57. Lukacs, N.W.; Moore, M.L.; Rudd, B.D.; Berlin, A.A.; Collins, R.D.; Olson, S.J.; Ho, S.B.; Peebles, R.S., Jr. Differential immune responses and pulmonary pathophysiology are induced by two different strains of respiratory syncytial virus. Am. J. Pathol. 2006, 169, 977–986. [Google Scholar] [CrossRef] [Green Version]
  58. Moore, M.L.; Chi, M.H.; Luongo, C.; Lukacs, N.W.; Polosukhin, V.V.; Huckabee, M.M.; Newcomb, D.C.; Buchholz, U.J.; Crowe, J.E., Jr.; Goleniewska, K.; et al. A chimeric A2 strain of respiratory syncytial virus (RSV) with the fusion protein of RSV strain line 19 exhibits enhanced viral load, mucus, and airway dysfunction. J. Virol. 2009, 83, 4185–4194. [Google Scholar] [CrossRef] [Green Version]
  59. Kim, Y.I.; DeVincenzo, J.P.; Jones, B.G.; Rudraraju, R.; Harrison, L.; Meyers, R.; Cehelsky, J.; Alvarez, R.; Hurwitz, J.L. Respiratory syncytial virus human experimental infection model: Provenance, production, and sequence of low-passaged memphis-37 challenge virus. PLoS ONE 2014, 9, e113100. [Google Scholar] [CrossRef] [Green Version]
  60. DeVincenzo, J.P.; Wilkinson, T.; Vaishnaw, A.; Cehelsky, J.; Meyers, R.; Nochur, S.; Harrison, L.; Meeking, P.; Mann, A.; Moane, E.; et al. Viral load drives disease in humans experimentally infected with respiratory syncytial virus. Am. J. Respir. Crit. Care Med. 2010, 182, 1305–1314. [Google Scholar] [CrossRef]
  61. DeVincenzo, J.; Lambkin-Williams, R.; Wilkinson, T.; Cehelsky, J.; Nochur, S.; Walsh, E.; Meyers, R.; Gollob, J.; Vaishnaw, A. A randomized, double-blind, placebo-controlled study of an RNAi-based therapy directed against respiratory syncytial virus. Proc. Natl. Acad. Sci. USA 2010, 107, 8800–8805. [Google Scholar] [CrossRef] [Green Version]
  62. Cantu-Flores, K.; Rivera-Alfaro, G.; Munoz-Escalante, J.C.; Noyola, D.E. Global distribution of respiratory syncytial virus A and B infections: A systematic review. Pathog. Glob. Health 2022, 116, 1–12. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, W.; Choi, Y.; Haynes, L.M.; Harcourt, J.L.; Anderson, L.J.; Jones, L.P.; Tripp, R.A. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bukreyev, A.; Yang, L.; Fricke, J.; Cheng, L.; Ward, J.M.; Murphy, B.R.; Collins, P.L. The secreted form of respiratory syncytial virus G glycoprotein helps the virus evade antibody-mediated restriction of replication by acting as an antigen decoy and through effects on Fc receptor-bearing leukocytes. J. Virol. 2008, 82, 12191–12204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Anderson, L.J.; Jadhao, S.J.; Paden, C.R.; Tong, S. Functional Features of the Respiratory Syncytial Virus G Protein. Viruses 2021, 13, 1214. [Google Scholar] [CrossRef]
  66. Van Royen, T.; Rossey, I.; Sedeyn, K.; Schepens, B.; Saelens, X. How RSV Proteins Join Forces to Overcome the Host Innate Immune Response. Viruses 2022, 14, 419. [Google Scholar] [CrossRef]
  67. Becker, Y. Respiratory syncytial virus (RSV) evades the human adaptive immune system by skewing the Th1/Th2 cytokine balance toward increased levels of Th2 cytokines and IgE, markers of allergy--a review. Virus Genes 2006, 33, 235–252. [Google Scholar] [CrossRef]
  68. Escribano-Romero, E.; Rawling, J.; Garcia-Barreno, B.; Melero, J.A. The soluble form of human respiratory syncytial virus attachment protein differs from the membrane-bound form in its oligomeric state but is still capable of binding to cell surface proteoglycans. J. Virol. 2004, 78, 3524–3532. [Google Scholar] [CrossRef] [Green Version]
  69. Arnold, R.; Konig, B.; Werchau, H.; Konig, W. Respiratory syncytial virus deficient in soluble G protein induced an increased proinflammatory response in human lung epithelial cells. Virology 2004, 330, 384–397. [Google Scholar] [CrossRef]
  70. Harcourt, J.; Alvarez, R.; Jones, L.P.; Henderson, C.; Anderson, L.J.; Tripp, R.A. Respiratory syncytial virus G protein and G protein CX3C motif adversely affect CX3CR1+ T cell responses. J. Immunol. 2006, 176, 1600–1608. [Google Scholar] [CrossRef]
  71. Johnson, C.H.; Miao, C.; Blanchard, E.G.; Caidi, H.; Radu, G.U.; Harcourt, J.L.; Haynes, L.M. Effect of chemokine receptor CX3CR1 deficiency in a murine model of respiratory syncytial virus infection. Comp. Med. 2012, 62, 14–20. [Google Scholar] [PubMed]
  72. Barik, S. Respiratory syncytial virus mechanisms to interfere with type 1 interferons. Curr. Top. Microbiol. Immunol. 2013, 372, 173–191. [Google Scholar] [CrossRef] [PubMed]
  73. Moore, E.C.; Barber, J.; Tripp, R.A. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hijano, D.R.; Vu, L.D.; Kauvar, L.M.; Tripp, R.A.; Polack, F.P.; Cormier, S.A. Role of Type I Interferon (IFN) in the Respiratory Syncytial Virus (RSV) Immune Response and Disease Severity. Front. Immunol. 2019, 10, 566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Taveras, J.; Garcia-Maurino, C.; Moore-Clingenpeel, M.; Xu, Z.; Mertz, S.; Ye, F.; Chen, P.; Cohen, S.H.; Cohen, D.; Peeples, M.E.; et al. Type-III Interferons, Viral Loads, Age, and Disease Severity in Young Children with Respiratory Syncytial Virus Infection. J. Infect. Dis. 2022, jiac404. [Google Scholar] [CrossRef]
  76. Selvaggi, C.; Pierangeli, A.; Fabiani, M.; Spano, L.; Nicolai, A.; Papoff, P.; Moretti, C.; Midulla, F.; Antonelli, G.; Scagnolari, C. Interferon lambda 1-3 expression in infants hospitalized for RSV or HRV associated bronchiolitis. J. Infect. 2014, 68, 467–477. [Google Scholar] [CrossRef]
  77. Blount, R.E., Jr.; Morris, J.A.; Savage, R.E. Recovery of cytopathogenic agent from chimpanzees with coryza. Proc. Soc. Exp. Biol. Med. 1956, 92, 544–549. [Google Scholar] [CrossRef]
  78. Miao, C.; Radu, G.U.; Caidi, H.; Tripp, R.A.; Anderson, L.J.; Haynes, L.M. 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. [Google Scholar] [CrossRef]
  79. Tripp, R.A.; Power, U.F.; Openshaw, P.J.M.; Kauvar, L.M. Respiratory Syncytial Virus: Targeting the G Protein Provides a New Approach for an Old Problem. J. Virol. 2018, 92, e01302-17. [Google Scholar] [CrossRef] [Green Version]
  80. Boyoglu-Barnum, S.; Todd, S.O.; Chirkova, T.; Barnum, T.R.; Gaston, K.A.; Haynes, L.M.; Tripp, R.A.; Moore, M.L.; Anderson, L.J. An anti-G protein monoclonal antibody treats RSV disease more effectively than an anti-F monoclonal antibody in BALB/c mice. Virology 2015, 483, 117–125. [Google Scholar] [CrossRef]
  81. Caidi, H.; Miao, C.; Thornburg, N.J.; Tripp, R.A.; Anderson, L.J.; Haynes, L.M. Anti-respiratory syncytial virus (RSV) G monoclonal antibodies reduce lung inflammation and viral lung titers when delivered therapeutically in a BALB/c mouse model. Antivir. Res. 2018, 154, 149–157. [Google Scholar] [CrossRef] [PubMed]
  82. Boyoglu-Barnum, S.; Chirkova, T.; Todd, S.O.; Barnum, T.R.; Gaston, K.A.; Jorquera, P.; Haynes, L.M.; Tripp, R.A.; Moore, M.L.; Anderson, L.J. Prophylaxis with a respiratory syncytial virus (RSV) anti-G protein monoclonal antibody shifts the adaptive immune response to RSV rA2-line19F infection from Th2 to Th1 in BALB/c mice. J. Virol. 2014, 88, 10569–10583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Collarini, E.J.; Lee, F.E.; Foord, O.; Park, M.; Sperinde, G.; Wu, H.; Harriman, W.D.; Carroll, S.F.; Ellsworth, S.L.; Anderson, L.J.; et al. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Haynes, L.M.; Caidi, H.; Radu, G.U.; Miao, C.; Harcourt, J.L.; Tripp, R.A.; Anderson, L.J. 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. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bergeron, H.C.; Tripp, R.A. RSV Replication, Transmission, and Disease Are Influenced by the RSV G Protein. Viruses 2022, 14, 2396. https://doi.org/10.3390/v14112396

AMA Style

Bergeron HC, Tripp RA. RSV Replication, Transmission, and Disease Are Influenced by the RSV G Protein. Viruses. 2022; 14(11):2396. https://doi.org/10.3390/v14112396

Chicago/Turabian Style

Bergeron, Harrison C., and Ralph A. Tripp. 2022. "RSV Replication, Transmission, and Disease Are Influenced by the RSV G Protein" Viruses 14, no. 11: 2396. https://doi.org/10.3390/v14112396

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop