Available online at www.sciencedirect.com
Pathogenesis of respiratory syncytial virus Sylvia van Drunen Littel-van den Hurk1,2 and Ellen R Watkiss1,3 While affecting all age groups, respiratory syncytial virus (RSV) infections can be particularly severe in infants, who develop functionally distinct immune responses, as well as in immunocompromised individuals. The extent to which environmental, viral and host factors contribute to the pathogenesis of RSV varies considerably between infected individuals. A correlation between the level of virus replication and pathogenesis has been established, and several viral proteins, in particular NS1 and NS2, modulate the immune response. Host immunity clearly contributes to RSV pathogenesis, and a number of specific cell populations may be involved. Ultimately, whether the response induced by RSV is protective or pathogenic depends on a combination of host factors, young age being one of the most important ones. Addresses 1 VIDO-Intervac, University of Saskatchewan, 120 Veterinary Rd., Saskatoon, SK, S7N 5E3, Canada 2 Microbiology and Immunology, University of Saskatchewan, 120 Veterinary Rd., Saskatoon, SK, S7N 5E3, Canada 3 Veterinary Microbiology, University of Saskatchewan, 120 Veterinary Rd., Saskatoon, SK, S7N 5E3, Canada Corresponding author: van Drunen Littel-van den Hurk, Sylvia (
[email protected])
Current Opinion in Virology 2012, 2:300–305 This review comes from a themed issue on Viral pathogenesis Edited by Diane Griffin and Veronika von Messling Available online 23rd February 2012 1879-6257/$ – see front matter # 2012 Elsevier B.V. All rights reserved. DOI 10.1016/j.coviro.2012.01.008
Introduction Respiratory syncytial virus (RSV) is a negative-strand, non-segmented RNA pneumovirus of the family Paramyxoviridae, and a common human pathogen that causes cold-like symptoms in most healthy adults and children. In infants and young children predisposed to respiratory illness, however, RSV infection is more likely to move into the lower respiratory tract (LRT), leading to pneumonia and bronchiolitis. RSV can be isolated from the majority of children hospitalized for bronchiolitis during the RSV season and is the most common respiratory virus in infants [1]. An increased incidence of asthma later in life has been associated with more severe LRT infections (LRTI) [2]. In older children, RSV is thought to be an important contributor to otitis media [3]. The winter and spring months of November through April are the peak Current Opinion in Virology 2012, 2:300–305
season of RSV infections in temperate climates of the Northern hemisphere, while in tropical climates RSV outbreaks occur most frequently during the rainy season [4,5]. RSV rapidly spreads through communities, infects most of the population within their first year of life, and re-infects individuals multiple times over the course of a lifetime (reviewed in [6]). In 2005, RSV caused 34 million cases of LRTI in children <5 years of age, 10% of whom required hospitalization [5]. While during the past few years RSVassociated mortality in developed countries decreased, probably owing to improved supportive care [6], the death rates are much higher in areas where access to healthcare is limited, such as in developing nations and remote areas of North America [7]. The mortality in 2005 was 66 000– 199 000 worldwide, of which 99% occurred in developing nations [5]. These numbers are probably underestimates [6]. RSV also is an important cause of mortality in elderly patients [8]. As the morbidity and mortality of infants, immunocompromised, and elderly individuals continue to rise worldwide, there still is a need for a better understanding of the mechanism of RSV pathogenesis.
Factors playing a role in RSV pathogenesis and disease outcome There are several schools of thought on what drives the pathogenesis of RSV, in particular the extent to which environmental, viral and host factors contribute to severe disease in infants (Figure 1). The immune-mediated nature of RSV pathology was highlighted by the failed vaccine that led to severe, non-protective, Th2-biased immunopathology upon RSV infection [9]. This, in combination with the striking association between severe bouts of RSV in infancy and the development of Th2mediated conditions like asthma and allergies in childhood, led many researchers to focus on the T-helper bias of the immune response to RSV in severe and mild infections. Others are more concerned with additional T cell populations or the innate responses produced in the lungs in the form of pro-inflammatory chemokines responsible for the influx of immune cells involved in pathogenesis. Environmental and social factors
Numerous factors may contribute to RSV disease severity. The age at the onset of the RSV season is a definite risk (<3 months or even <6 months). The presence of smoking members or young siblings in the household, daycare attendance, stay in a nursing home or hospital also increase the risk of exposure and serious infection with RSV (reviewed in [10]). www.sciencedirect.com
RSV pathogenesis van Drunen Littel-van den Hurk and Watkiss 301
Figure 1
Viral factors
Environmental factors
•Viral load and isolate •Down-regulation of type I IFN response by NS1 and NS2 •Escape from neutralization (sG) •Reduction fraktalkine action (G)
•Smoking in household •Young siblings •Daycare attendance •Stay in a hospital
RSV infection of epithelial cells
Virus replication and cytopathology
Immunopathology
Genetic polymorphisms •Innate defense genes Sloughing of cells •Surfactant protein genes Reduced ciliary funcion •Host cell receptor genes •Neutrophil response genes •Th1/Th2 response genes •Gene effectors of adaptive immunity
Cell infiltration Mucus production
Host factors •Premature birth and low birth weight •Chronic lung disease of prematurity • Congenital heart disease •Incomplete development, damage or hyperreactivity of the airway •Immunodeficiency •Male gender •Multiple births •Low titer of RSV-specific maternal antibodies •Cord blood vitamin D deficiency
Bronchiolitis Airway insufficiency Current Opinion in Virology
Factors influencing the pathogenesis and clinical disease caused by RSV infection in infants and young children.
Exposure to smoke, whether from air pollution or cigarettes, has a detrimental effect on respiratory health in general. According to a recent study the risk for development of RSV broncheolitis is increased by trafficderived pollution [11]. A more definite link was shown between smoking in the household, and the incidence and severity of RSV infection [12]. Furthermore, in an ex vivo tissue culture system of primary airway epithelial cells cigarette smoke caused necrosis rather than virusinduced apoptosis resulting in increased inflammation and enhanced viral replication; this effect was primarily mediated by reactive aldehydes [13]. Viral factors
RSV is not a highly cytopathic virus. RSV targets both type I alveolar and nonbasilar airway epithelial cells and possibly alveolar macrophages [14]. This results in impairment of the ciliary action and sloughing of infected epithelial cells. Peribroncheal mononuclear cell infiltration, submucosal edema, mucus secretion, and sometimes syncytia are observed in the lung. When polarized, mucociliary epithelial cells were infected with RSV in vitro, replication was limited to the apical surface [15]. RSV www.sciencedirect.com
does not induce general shut-off of host cell transcription; however, changes in abundance of cell-cycle-regulatory proteins were observed in primary HBEpC cells as well as A549 cells, leading to enrichment in the G0/G1 population and increased progeny virus [16]. Despite its low cytopathicity, there is compelling evidence that the level of RSV replication correlates to the disease severity [17,18]. Furthermore, while evidence that RSV subgroup A (or an A genotype) is more virulent has been controversial, subgroup A strain 19 was recently shown to induce increased production of IL-13, mucus production and airway hypersensitivity in mice [19]. Subsequently, RSV clinical isolates were found to induce variable pathogenesis. Several RSV subgroup A isolates had similar growth properties in BEAS-2B cells, but one of them, RSV A2001/2-20, caused greater disease severity, higher lung IL-13 levels, and higher lung gob-5 levels than RSV strains A2, line 19, Long, and A2001/3-12 in BALB/cJ mice. In addition, the A2001/2-20 clinical isolate induced airway mucin expression, which was correlated to F protein characteristics. Two of the clinical isolates, RSV 2-20 and A2001/3-12, replicated to higher titers in the Current Opinion in Virology 2012, 2:300–305
302 Viral pathogenesis
lung than laboratory RSV strains on day 1, which correlated with greater histopathologic changes. RSV 2-20 induced epithelial desquamation, bronchiolitis, airway hyperresponsiveness, and increased breathing effort [20]. Several RSV proteins play a role in modulation of the host response to infection. The NS1 and NS2 proteins block IFN regulatory factor 3 activation and inhibit the type I IFN induced signaling through the JAK/STAT pathway leading to a very effective block of IFNa/b production by the infected host. RSV further downregulates IFN production and function by inhibiting toll-like receptor (TLR) signaling through myeloid differentiation factor 88 and mitochondrial antiviral signaling protein [21], as well as induction of suppressor of cytokine signaling molecules [22] and interference with RIG-I [23]. In addition, NS1 and NS2 activate the phosphoinositidide 3-kinase pathway, which results in reduced apoptosis and thus enhanced survival of the infected cells [24]. The G protein plays a major role in immune evasion of RSV. First, it is a highly glycosylated protein, which may impede immune recognition. It also is highly variable, with only 53% amino acid identity and 1–7% antigenic relatedness between subgroups, which allows easy escape from neutralizing antibodies. Furthermore, in addition to the full-length G protein a truncated, secreted form, sG, is produced during RSV infection at 4-fold the rates present in progeny virus. This sG binds RSV-specific antibodies and thus reduces the concentrations available for RSV neutralization [25]. The G protein also has limited sequence homology to fraktalkine, and can reduce the action of host fraktalkine and the influx of CXCR1 leukocytes such as natural killer (NK) cells, and CD4 and CD8 T cells [26]. Finally, the sG protein functions as a TLR antagonist, and down-regulates the TLR2-, TLR4- and TLR9-mediated inflammatory response [27]. RSV also has the ability to infect different populations of immune cells, including macrophages and dendritic cells (DCs), affecting their antigen presenting capacity (reviewed in [28]). It is not surprising that such effects may be more prominent in neonates, which are immature at the level of DC development. Recently, a role for NS1 was found in suppression of CD103+CD8+ T cells and Th17 cells, and stimulation of Th2 cells. This was probably owing to NS1 causing reduced maturation of DCs [29]. Interestingly, ex vivo RSV-stimulated monocytederived DCs suboptimally upregulated the CCR7 receptor and thus migrated less efficiently to CCL19 than for example influenza virus. As the CCR7 receptor is critical for migration of DCs to the lymphoid tissues, this may contribute to the reduced adaptive immune responses to RSV [30]. This is in agreement with a previous observation that in children and mice, DCs persisted in the respiratory mucosa and secretions for weeks after the acute symptoms of RSV infection had resolved [31,32]. Current Opinion in Virology 2012, 2:300–305
Another cell population, human bone marrow stromal cells (BMSCs), was recently identified as a novel target for RSV, which induced disruption of cytoskeletal actin filaments, altered chemokine/cytokine expression, decreased ability to stimulate B cell maturation, and modulated chemotaxis. RNA sequences homologous to the RSV genome were found in naive primary human BMSCs. This may have consequences for the acute RSV induced pathogenesis as well as long-term effects [33]. Host factors
Very little is known about the normal host response to RSV infection, so much of the information about the impact of host factors is based on LRTI. More severe LRT RSV infections are correlated to premature birth, chronic lung disease of prematurity, congenital heart disease, and T-cell immunodeficiency. Incomplete development or damage to the airway, and/or airway hyperreactivity of premature infants contribute to RSV-induced morbidity. Immunodeficiency, immunosuppression or old age may lead to prolonged viral replication and more severe illness. Other risk factors are male gender, low birth weight, multiple births and low titer of RSV-specific maternal antibodies (discussed in [34]). Interestingly, recent evidence suggests that cord blood vitamin D deficiency in healthy neonates is associated with increased risk of severe RSV LRTI in the first year of life [35]. Several genetic polymorphisms, including genes involved in the innate defense, surfactant protein genes, host cell receptor genes, neutrophil response genes, Th1/Th2 response genes and gene effectors of adaptive immunity, have been reported. Although the genetic factors related to RSV severity were suggested to be probably complex and polygeneic, certain loci affecting the ability of the host to control early viral replication as well as candidate loci affecting potential immunopathology, were identified as significant factors in the severity of human RSV disease (reviewed in [36]). The importance of epithelial cells and macrophages in the innate immune response to RSV was recently recognized. Several chemokines and cytokines including IL-8/ CXCL8, IP-10/CXCL10, MCP-1/CCL2, MIP-1a/CCL3, MIP-1b/CCL4, RANTES/CCL5, IL-6, TNF-a, IL-1ab, and IFN-a/b are produced by epithelial cells and macrophages in response to RSV infection, and found in enhanced amounts in respiratory secretions of children hospitalized for RSV infection [37]. Upregulation of IL8 is correlated to the severity of RSV disease [38], and leads to recruitment of neutrophils, which constitute the majority of infiltrating cells (at least 84%) [39]. While neutrophils may mediate elimination of virus-infected cells, their high numbers, ability to secrete further cytokines and chemokines, and degranulation products may contribute to RSV-induced immunopathogenesis. Indeed, instead of eosinophils as originally proposed, neutrophils www.sciencedirect.com
RSV pathogenesis van Drunen Littel-van den Hurk and Watkiss 303
constituted the predominant population in FI-RSV vaccinated children who died of subsequent RSV infection. Moreover, although increased eosinophil degranulation products were found in the respiratory secretions of children with RSV bronchiolitis [40], eosinophils represent only 1–3% of the airway leukocytes in RSV infected infants [39], and in mice can be protective [41]. Interestingly, an inverse correlation between IL-8 concentrations in the amniotic fluid and wheezing was found, suggesting that inflammatory cytokines in the amniotic fluid accelerate maturation of the fetal lung, which promotes protection from severe RSV infection [42]. While the RSV-specific T cell response plays a major role in viral clearance and the clinical outcome of infection, both Th2-biased CD4+ and CD8+ T cells have been implicated in immunopathogenesis (reviewed in [6,43]). Recently, regulatory T cells (Tregs), were shown to also play a critical role in regulating the innate and adaptive responses during the later stages of RSV lung infection; depletion of Tregs before RSV infection resulted in delayed viral clearance and increased disease severity in mice [44,45,46]. While in one study innate immune cells in lung and bronchoalveolar lavage (BAL), as well influx of CD4+ and CD8+ IFN-g producing T cells were increased [45], a delay in early CD8+ T cell recruitment into the lung [44,46], in one case followed by an increase at a later time point [44] was reported by other groups. Enhanced lung cytokine and chemokine levels, and increased NK cells and neutrophils in the BAL, were also observed [45]. As IL-17 is known to play a role in the development of asthma, its role in RSV pathogenesis was recently examined. Increased IL-6 and IL-17 levels were found in the tracheal aspirate samples from severely ill RSV-infected infants. Furthermore, IL-6, IL-17 and IL-23 were increased in RSV-infected mice, while treatment with anti-IL17 antibodies reduced inflammation, decreased viral load, and increased antigenspecific CD8+ T cells in the lung [47]. This study agrees with the recently reported detection of numerous IL-17positive cells in the lung sections of children who died of enhanced RSV disease in 1967 [42].
While the mouse model has advantages, in particular the availability of reagents and knockout strains, the host specificity of RSV makes studies in mice, as well as cotton rats, difficult to interpret in the context of human infections. Another limiting factor in most RSV studies is the use of adult mice. RSV-infected neonatal mice (7 days of age) were shown to develop long-term asthma-like disease including increased airway hypersensitivity, mucus hyperproduction, airway remodeling, and Th2 cytokines and cellular responses, which makes this a potentially more attractive model (reviewed in [48]). Alternative animal models include pneumonia virus of mice. As a natural pathogen in rodents, a low-dose inoculum replicates to high levels and causes immunemediated respiratory illness that is quite similar to that described for RSV in children (reviewed in [49]). Bovine RSV (BRSV) infection of calves constitutes another appropriate model, as the pathologic lesions caused by human and bovine RSV are very similar (reviewed in [50]). Furthermore, as for RSV, age-dependent differences in BRSV-induced pathogenesis were recently demonstrated [51]. However, the expense of using calves as a model is a limiting factor. This applies even more so to non-human primates, even though they are a natural target for RSV. Recently, a newborn lamb model was established, both for human and bovine RSV [52]. The pathology of RSV in lambs is similar to that in human infants, that is, mild peribronchiolar infiltrates of lymphocytes and plasma cells, and bronchiolitis characterized by degeneration and sloughing of epithelial cells, and intraluminal infiltrates of neutrophils [52]. The airway structure and function of newborn lambs are more similar to those in infants compared to mice [53]. Furthermore, alveolar development occurs preterm in both humans and lambs [54], and pre-term lambs can be examined for their ability to respond to RSV infection [55], making this an attractive model to study RSV pathogenesis.
Acknowledgements Animal models of pathogenesis Studying the pathogenesis of RSV in humans is difficult for a number of reasons. Many RSV infections in healthy individuals are mild enough to remain untreated and unreported, so often only the most severe infections requiring medical intervention can be studied. Also, the site of infection in the LRT is not readily accessible, limiting studies to non-invasive procedures such as the analysis of peripheral blood cells, nasal washes, lung aspirates from intubated infants, and post-mortem lung tissue. The availability of appropriate control samples and the ethical implications of sampling the LRT in severely ill infants pose further limits. Animal models of RSV infection overcome these limitations by providing access to the site of inflammation. www.sciencedirect.com
The contributions of all current and previous members of the laboratory, colleagues and the animal care group at VIDO-Intervac are gratefully acknowledged. Work in the author’s laboratory is currently supported by the Natural Sciences and Engineering Research Council of Canada, Canadian Institutes of Health Research, Krembil Foundation, Saskatchewan Health Research Foundation, Agricultural Development Fund of Saskatchewan, and Alberta Livestock and Meat Agency.
References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as: of special interest of outstanding interest 1.
Hall CB, Weinberg GA, Iwane MK, Blumkin AK, Edwards KM, Staat MA, Auinger P, Griffin MR, Poehling KA, Erdman D et al.: The burden of respiratory syncytial virus infection in young children. N Engl J Med 2009, 360:588-598. Current Opinion in Virology 2012, 2:300–305
304 Viral pathogenesis
2.
Mailaparambil B, Grychtol R, Heinzmann A: Respiratory syncytial virus bronchiolitis and asthma – insights from recent studies and implications for therapy. Inflamm Allergy Drug Targets 2009, 8:202-207.
3.
Gomaa MA, Galal O, Mahmoud MS: Risk of acute otitis media in relation to acute bronchiolitis in children. Int J Pediatr Otorhinolaryngol 2011, 76:49-51.
4.
Centers for Disease Control and Prevention (CDC): Respiratory syncytial virus – United States, July 2007–June 2011. MMWR Morb Mortal Wkly Rep 2011, 60:1203–1206.
5.
Nair H, Nokes DJ, Gessner BD, Dherani M, Madhi SA, Singleton RJ, O’Brien KL, Roca A, Wright PF, Bruce N et al.: Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet 2010, 375:1545-1555.
6.
Collins PL, Melero JA: Progress in understanding and controlling respiratory syncytial virus: still crazy after all these years. Virus Res 2011, 162:80-99.
7.
Lowther SA, Shay DK, Holman RC, Clarke MJ, Kaufman SF, Anderson LJ: Bronchiolitis-associated hospitalizations among American Indian and Alaska Native children. Pediatr Infect Dis J 2000, 19:11-17.
8.
Falsey AR, Walsh EE: Respiratory syncytial virus infection in elderly adults. Drugs Aging 2005, 22:577-587.
9.
Kim HW, Canchola JG, Brandt CD, Pyles G, Chanock RM, Jensen K, Parrott RH: Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol 1969, 89:422-434.
10. Committee on Infectious Diseases: From the American Academy of Pediatrics: Policy statements – modified recommendations for use of palivizumab for prevention of respiratory syncytial virus infections. Pediatrics 2009, 124:1694–1701. 11. Karr CJ, Rudra CB, Miller KA, Gould TR, Larson T, Sathyanarayana S, Koenig JQ: Infant exposure to fine particulate matter and traffic and risk of hospitalization for RSV bronchiolitis in a region with lower ambient air pollution. Environ Res 2009, 109:321-327. 12. Bradley JP, Bacharier LB, Bonfiglio J, Schechtman KB, Strunk R, Storch G, Castro M: Severity of respiratory syncytial virus bronchiolitis is affected by cigarette smoke exposure and atopy. Pediatrics 2005, 115:e7-e14. 13. Groskreutz DJ, Monick MM, Babor EC, Nyunoya T, Varga SM, Look DC, Hunninghake GW: Cigarette smoke alters respiratory syncytial virus-induced apoptosis and replication. Am J Respir Cell Mol Biol 2009, 41:189-198. 14. Johnson JE, Gonzales RA, Olson SJ, Wright PF, Graham BS: The histopathology of fatal untreated human respiratory syncytial virus infection. Mod Pathol 2007, 20:108-119. 15. Wright PF, Ikizler MR, Gonzales RA, Carroll KN, Johnson JE, Werkhaven JA: Growth of respiratory syncytial virus in primary epithelial cells from the human respiratory tract. J Virol 2005, 79:8651-8654. 16. Wu W, Munday DC, Howell G, Platt G, Barr JN, Hiscox JA: Characterization of the interaction between human respiratory syncytial virus and the cell cycle in continuous cell culture and primary human airway epithelial cells. J Virol 2011, 85:10300-10309. 17. DeVincenzo JP, 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. 18. El Saleeby CM, Bush AJ, Harrison LM, Aitken JA, Devincenzo JP: Respiratory syncytial virus load, viral dynamics, and disease severity in previously healthy naturally infected children. J Infect Dis 2011, 204:996-1002. A positive correlation between duration of clinical disease and RSV viral load was demonstrated. These observations challenge the immunopathology-based pathogenesis paradigm, and suggest that (early) treatment with antiviral agents might improve subsequent morbidity. Current Opinion in Virology 2012, 2:300–305
19. Moore ML, Chi MH, Luongo C, Lukacs NW, Polosukhin VV, Huckabee MM, Newcomb DC, Buchholz UJ, Crowe JE 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. 20. Stokes KL, Chi MH, Sakamoto K, Newcomb DC, Currier MG, Huckabee MM, Lee S, Goleniewska K, Pretto C, Williams JV et al.: Differential pathogenesis of respiratory syncytial virus clinical isolates in BALB/c mice. J Virol 2011, 85:5782-5793. Several clinical isolates were tested and shown to induce variable pathogenesis in BALB/c mice. A mouse model that shows characteristic RSV pathogenesis was developed with one of these isolates. 21. Bhoj VG, Sun Q, Bhoj EJ, Somers C, Chen X, Torres JP, Mejias A, Gomez AM, Jafri H, Ramilo O et al.: MAVS and MyD88 are essential for innate immunity but not cytotoxic T lymphocyte response against respiratory syncytial virus. Proc Natl Acad Sci USA 2008, 105:14046-14051. 22. Hashimoto K, Ishibashi K, Ishioka K, Zhao D, Sato M, Ohara S, Abe Y, Kawasaki Y, Sato Y, Yokota S et al.: RSV replication is attenuated by counteracting expression of the suppressor of cytokine signaling (SOCS) molecules. Virology 2009, 391:162-170. 23. Ling Z, Tran KC, Teng MN: Human respiratory syncytial virus nonstructural protein NS2 antagonizes the activation of beta interferon transcription by interacting with RIG-I. J Virol 2009, 83:3734-3742. 24. Bitko V, Shulyayeva O, Mazumder B, Musiyenko A, Ramaswamy M, Look DC, Barik S: Nonstructural proteins of respiratory syncytial virus suppress premature apoptosis by an NF-kappaB-dependent, interferon-independent mechanism and facilitate virus growth. J Virol 2007, 81:1786-1795. 25. Bukreyev A, Yang L, Fricke J, Cheng L, Ward JM, Murphy BR, Collins PL: 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. 26. 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. 27. Polack FP, Irusta PM, Hoffman SJ, Schiatti MP, Melendi GA, Delgado MF, Laham FR, Thumar B, Hendry RM, Melero JA et al.: The cysteine-rich region of respiratory syncytial virus attachment protein inhibits innate immunity elicited by the virus and endotoxin. Proc Natl Acad Sci USA 2005, 102:8996-9001. 28. Lukacs NW, Smit JJ, Schaller MA, Lindell DM: Regulation of immunity to respiratory syncytial virus by dendritic cells, tolllike receptors, and notch. Viral Immunol 2008, 21:115-122. 29. Munir S, Hillyer P, Le Nouen C, Buchholz UJ, Rabin RL, Collins PL, Bukreyev A: Respiratory syncytial virus interferon antagonist NS1 protein suppresses and skews the human T lymphocyte response. PLoS Pathogens 2011, 7:e1001336. NS1 protein was shown to suppress proliferation and activation of CD103+ CD8+ T cells and Th17 cells, and to promote proliferation and activation of Th2 cells. Evidence for downregulation of maturation by NS1 and NS2 was provided. 30. Le Nouen C, Hillyer P, Winter CC, McCarty T, Rabin RL, Collins PL, Buchholz UJ: Low CCR7-mediated migration of human monocyte derived dendritic cells in response to human respiratory syncytial virus and human metapneumovirus. PLoS Pathogens 2011, 7:e1002105. RSV infection of human monocyte-derived dendritic cells (mo-DCs) led to reduced increase in CCR7 expression, and mo-DCs stimulated with RSV migrated inefficiently to CCL19. This suggests that impaired migration of RSV-stimulated DCs to lymphatic tissue contributes to impaired adaptive responses to RSV. 31. Beyer M, Bartz H, Horner K, Doths S, Koerner-Rettberg C, Schwarze J: Sustained increases in numbers of pulmonary dendritic cells after respiratory syncytial virus infection. J Allergy Clin Immunol 2004, 113:127-133. www.sciencedirect.com
RSV pathogenesis van Drunen Littel-van den Hurk and Watkiss 305
32. Gill MA, Palucka AK, Barton T, Ghaffar F, Jafri H, Banchereau J, Ramilo O: Mobilization of plasmacytoid and myeloid dendritic cells to mucosal sites in children with respiratory syncytial virus and other viral respiratory infections. J Infect Dis 2005, 191:1105-1115. 33. Rezaee F, Gibson LF, Piktel D, Othumpangat S, Piedimonte G: Respiratory syncytial virus infection in human bone marrow stromal cells. Am J Respir Cell Mol Biol 2011, 45:277-286. Human bone marrow stromal cells (BMSCs) were identified as a novel target for RSV, both in vivo and ex vivo. RSV infection of BMSCs resulted in structural and functional changes, which may influence the pathogenesis. 34. Groothuis JR, Hoopes JM, Jessie VG: Prevention of serious respiratory syncytial virus-related illness. I: Disease pathogenesis and early attempts at prevention. Adv Ther 2011, 28:91-109. 35. Belderbos ME, Houben ML, Wilbrink B, Lentjes E, Bloemen EM, Kimpen JL, Rovers M, Bont L: Cord blood vitamin D deficiency is associated with respiratory syncytial virus bronchiolitis. Pediatrics 2011, 127:e1513-e1520. An analysis of cord blood vitamin D levels showed that vitamin D deficiency is correlated with increased risk of RSV LRTI in the first year of life This suggests vitamin D supplementation during pregnancy as a strategy to prevent RSV LRTI.
45. Lee DC, Harker JA, Tregoning JS, Atabani SF, Johansson C, Schwarze J, Openshaw PJ: CD25+ natural regulatory T cells are critical in limiting innate and adaptive immunity and resolving disease following respiratory syncytial virus infection. J Virol 2010, 84:8790-8798. In increase in natural Treg numbers in the bronchoalveolar lavage fluid, lung, mediastinal lymph nodes, and spleen was demonstrated in RSVinfected mice. Depletion of Tregs led to increased weight loss. The Tregs were shown to play a critical role in regulating both innate and adaptive responses late during RSV infection. 46. Ruckwardt TJ, Bonaparte KL, Nason MC, Graham BS: Regulatory T cells promote early influx of CD8+ T cells in the lungs of respiratory syncytial virus-infected mice and diminish immunodominance disparities. J Virol 2009, 83:3019-3028. 47. Mukherjee S, Lindell DM, Berlin AA, Morris SB, Shanley TP, Hershenson MB, Lukacs NW: IL-17-induced pulmonary pathogenesis during respiratory viral infection and exacerbation of allergic disease. Am J Pathol 2011, 179:248-258. Increased IL-6 and IL-17 levels were detected in tracheal aspirate samples from severely ill infants with RSV infection. A role for IL-17 in RSV pathogenesis was subsequently established in the mouse model.
36. Miyairi I, DeVincenzo JP: Human genetic factors and respiratory syncytial virus disease severity. Clin Microbiol Rev 2008, 21:686-703.
48. Cormier SA, You D, Honnegowda S: The use of a neonatal mouse model to study respiratory syncytial virus infections. Expert Rev Anti-infective Ther 2010, 8:1371-1380.
37. McNamara PS, Flanagan BF, Hart CA, Smyth RL: Production of chemokines in the lungs of infants with severe respiratory syncytial virus bronchiolitis. J Infect Dis 2005, 191:1225-1232.
49. Rosenberg HF, Domachowske JB: Pneumonia virus of mice: severe respiratory infection in a natural host. Immunol Lett 2008, 118:6-12.
38. Goetghebuer T, Isles K, Moore C, Thomson A, Kwiatkowski D, Hull J: Genetic predisposition to wheeze following respiratory syncytial virus bronchiolitis. Clin Exp Allergy 2004, 34:801-803.
50. Gershwin LJ: Bovine respiratory syncytial virus infection: immunopathogenic mechanisms. Anim Health Res Rev 2007, 8:207-213.
39. McNamara PS, Ritson P, Selby A, Hart CA, Smyth RL: Bronchoalveolar lavage cellularity in infants with severe respiratory syncytial virus bronchiolitis. Arch Dis Child 2003, 88:922-926.
51. Antonis AF, de Jong MC, van der Poel WH, van der Most RG, Stockhofe-Zurwieden N, Kimman T, Schrijver RS: Agedependent differences in the pathogenesis of bovine respiratory syncytial virus infections related to the development of natural immunocompetence. J Gen Virol 2011, 91(Pt 10):2497-2506.
40. Harrison AM, Bonville CA, Rosenberg HF, Domachowske JB: Respiratory syncytical virus-induced chemokine expression in the lower airways: eosinophil recruitment and degranulation. Am J Respir Crit Care Med 1999, 159:1918-1924. 41. Phipps S, Lam CE, Mahalingam S, Newhouse M, Ramirez R, Rosenberg HF, Foster PS, Matthaei KI: Eosinophils contribute to innate antiviral immunity and promote clearance of respiratory syncytial virus. Blood 2007, 110:1578-1586. 42. van Bleek GM, Osterhaus AD, de Swart RL: RSV 2010: recent advances in research on respiratory syncytial virus and other pneumoviruses. Vaccine 2011, 29:7285-7291. 43. van Drunen Littel-van den Hurk S, Mapletoft JW, Arsic N, KovacsNolan J: Immunopathology of RSV infection: prospects for developing vaccines without this complication. Rev Med Virol 2007, 17:5-34. 44. Fulton RB, Meyerholz DK, Varga SM: Foxp3+ CD4 regulatory T cells limit pulmonary immunopathology by modulating the CD8 T cell response during respiratory syncytial virus infection. J Immunol 2010, 185:2382-2392. A rapid accumulation of regulatory T-cells (Tregs) in the mediastinal lymph nodes and lungs of RSV infected mice was shown. In vivo depletion of Tregs before RSV infection led to delayed virus clearance and enhanced disease. This indicates that pulmonary Tregs play a critical role in limiting immunopathology during acute RSV infection.
www.sciencedirect.com
52. Sow FB, Gallup JM, Olivier A, Krishnan S, Patera AC, Suzich J, Ackermann MR: Respiratory syncytial virus is associated with an inflammatory response in lungs and architectural remodeling of lung-draining lymph nodes of newborn lambs. Am J Physiol Lung Cell Mol Physiol 2011, 300:L12-L24. A detailed characterization of the pathology induced by RSV A2 in newborn lambs demonstrated moderate clinical disease with increased virus titers, accumulation of macrophages, CD4+ and CD8+ T cells, and a transcriptional response of genes involved in inflammation, chemotaxis, and interferon response. 53. Scheerlinck JP, Snibson KJ, Bowles VM, Sutton P: Biomedical applications of sheep models: from asthma to vaccines. Trends Biotechnol 2008, 26:259-266. 54. Flecknoe SJ, Wallace MJ, Cock ML, Harding R, Hooper SB: Changes in alveolar epithelial cell proportions during fetal and postnatal development in sheep. Am J Physiol Lung Cell Mol Physiol 2003, 285:L664-L670. 55. Meyerholz DK, Grubor B, Fach SJ, Sacco RE, Lehmkuhl HD, Gallup JM, Ackermann MR: Reduced clearance of respiratory syncytial virus infection in a preterm lamb model. Microbes Infect 2004, 6:1312-1319.
Current Opinion in Virology 2012, 2:300–305