Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2008 Jul;21(3):495–504. doi: 10.1128/CMR.00054-07

Epidemiologic, Experimental, and Clinical Links between Respiratory Syncytial Virus Infection and Asthma

Shyam S Mohapatra 1,*, Sandhya Boyapalle 1
PMCID: PMC2493089  PMID: 18625684

Abstract

Virtually all children experience respiratory syncytial virus (RSV) infection at least once during the first 2 years of life, but only a few develop bronchiolitis and more severe disease requiring hospitalization, usually in the first 6 months of life. Children who recover from RSV-induced bronchiolitis are at increased risk for the development of recurrent wheeze and asthma in later childhood. Recent studies suggest that there is an association between RSV-induced bronchiolitis and asthma within the first decade of life but that this association is not significant after age 13. Despite the considerable progress made in our understanding of several aspects of respiratory viral infections, further work needs to be done to clarify the molecular mechanisms of early interactions between virus and host cell and the role of host gene products in the infection process. This review provides a critical appraisal of the literature in epidemiology and experimental research which links RSV infection to asthma. Studies to date demonstrate that there is a significant association between RSV infection and childhood asthma and that preventing severe primary RSV infections can decrease the risk of childhood asthma.

INTRODUCTION

Respiratory syncytial virus (RSV), the most common cause of severe lower respiratory tract disease among infants and young children, typically infects persons by 2 years of age and can cause subsequent infections throughout life (122). By 2 years of age, almost all children have been infected with RSV, and over half have been infected twice (38). In Spain, the annual hospitalization rate is 37 out of 1,000 infants under 6 months and out of 1,000 among those under 1 year of age (137). Persons at increased risk for severe disease or death related to RSV include premature infants, elderly adults, and persons of any age with compromised respiratory, cardiac, or immune systems (27, 139). RSV is transmitted from person to person via close contact, droplets, or fomites. In temperate climates, peak RSV activity typically occurs during the winter. However, year-to-year national and regional variability in the RSV season onset and offset occurs in the United States (87). RSV circulations also differ by geographic location; for example, Florida has an earlier season onset and a longer season than the rest of the United States (16). A case-control study was undertaken in southern Israel, and the authors examined the possible association between birth season (date of birth) and future development of asthma in children. Infants who were exposed to RSV infection at a very young age were better protected against the development of asthma, although those born at the end of winter and in early spring and who were exposed to RSV infection during the last quarter of their first year of life might be a higher risk for future development of asthma (36).

About 30% of children hospitalized for acute bronchiolitis will have recurrent respiratory symptoms in the years following their having the disease (124, 125, 127). This observation raises the possibility of a relationship between acute bronchiolitis and the subsequent development of bronchial hyperreactivity. A number of authors have studied a possible role of this virus in the development of asthma, although the nature of the association between bronchiolitis and asthma is not completely understood. Asthma could be a direct consequence of RSV infection itself, or the virus may trigger changes in pulmonary physiology in patients who are especially predisposed to asthma (117). RSV infection could increase susceptibility to asthma by acting on the immune response, genetic factors, or neural control of the respiratory tract (82). Experimental models have also shown long-term persistence of RSV in respiratory epithelial cells (121). The elucidation of the relationship between RSV infection and the development of asthma, as well as a better understanding of the nature of their association, would have important implications for prevention and treatment. However, the studies so far have been observational, and they differ widely with regard to population, design, methodology, and length of follow-up. A critical appraisal of the epidemiologic, experimental, and clinical links should enable us to better understand the relation between RSV infection and the development of asthma.

PATHOBIOLOGY OF RSV INFECTION

The RSV genome is composed of single-stranded negative-sense RNA. The mature, infectious virus particle consists of ribonucleoprotein (RNP) formed by the interaction of the viral genomic RNA with the nucleocapsid (N) protein, the phosphoprotein (P), and the large (L) protein. The M2-1 protein is a virus protein required for efficient transcription of the virus genome by the polymerase complex (20, 28, 45, 53, 142). The RNP is surrounded by two layers (the matrix [M] protein and a lipid envelope derived from host cell); three virus-encoded proteins (the attachment [G], fusion [F], and small hydrophobic [SH] proteins) are embedded within the envelope (3, 5, 15, 89, 96, 114). The two nonstructural proteins, NS1 and NS2, are not readily detectable in virions but are found in RSV-infected cells (26, 59).

The receptor(s) for RSV has not been unequivocally identified, but in vitro data provide some clues about the mechanisms of RSV entry. Heparin-like glycosaminoglycans, unbranched polysaccharide chains on the surface of most mammalian cells, have been implicated as receptors or coreceptors for RSV (78) or in playing some role in virus entry (52). Although glycosaminoglycans are important for RSV G protein interaction with target cells, the precise cellular receptor of RSV remains unknown. The viral F protein, which mediates viral penetration by fusion of the viral envelope with the host cell plasma membrane, has been shown to bind to intercellular adhesion molecule-1 (ICAM-1) (8, 29, 72, 75). Studies using both light and electron microscopy techniques have shown that RSV matures predominantly as filamentous structures on the surface of infected cells. Moreover, lipid rafts have been implicated in the assembly process of the virus particles (13-15, 62, 80).

The airway epithelium is the main target of RSV infection. After infection, RSV replicates in the respiratory mucosa, leading to epithelial damage and perivascular mononuclear infiltration (4, 30). When infected by virus, epithelial cells respond by producing a number of potent immunomodulatory and inflammatory mediators, including cytokines and chemokines (7, 31, 33, 88, 144). RSV infection activates signaling pathways in airway epithelial cells through the surface molecule toll-like receptor 4 (72). It was shown previously that RSV replication activates various transcription factors, including NF-κB, a central mediator of RSV-induced airway inflammation (33, 48, 61, 135, 144). The cytokines induced by viral infection are also known to activate the JAK/STAT (signal transducer and activator of transcription) signal transduction pathway, which might regulate the subsequent adaptive immune response (130). Subsequent to the acute infection, epithelial cells are potentially capable of presenting viral antigens to lymphoid cells (102, 103). Viral clearance involves the induction of both cellular and humoral immunity; however, immunity is considered incomplete, as the virus can reinfect the host (40, 54). Natural immunity to RSV appears to be minimal, and annual reinfections are frequent during the first years of life (57).

Autopsy studies of children who died of RSV-induced bronchiolitis revealed the degree of inflammation generated by the immune response and the subsequent airway obstruction. The infection results in the loss of cilia, sloughing of epithelial cells into the airway, collection of desquamated epithelial cells, polymorphonuclear leukocytes, fibrin, lymphocytes, and mucus within the airway, and edema around the airway. The degree of epithelial damage has been correlated with the magnitude of inflammation and airway hyperreactivity (AHR) (47, 54, 79, 105). Lower respiratory tract RSV infection in infancy significantly increased the odds of having wheezing fits up to 11 years of age in an outpatient population and more than quintupled the risk of asthma/recurrent wheeze at age 13 for infants who had been hospitalized with RSV bronchiolitis, in comparison to control subjects without infection (43% versus 8%) (127, 131).

EPIDEMIOLOGY OF RSV

RSV is the leading cause of bronchiolitis in infants worldwide. In temperate climates, most RSV infections occur between November and May, whereas in tropical climates, RSV infections occur year-round (12). In the United States, RSV-related disease is a yearly epidemic that peaks in January or February in most parts of the country and slightly earlier in the southeast (17). During the winter epidemics, RSV can infect up to 100% of the children in day care centers (56). The age related to the highest morbidity risk of RSV infection is (50, 95). RSV is also a major cause of respiratory illness in the elderly and high-risk adults. It has been estimated that more than 120,000 infants in the United States will be hospitalized annually with RSV infection, with more than 200 deaths occurring as a result of this illness (122, 123). About 50% of infants hospitalized for lower respiratory tract RSV infection have subsequent episodes of wheezing that in some cases can persist to 11 years of age or later ages (51, 129). There is also evidence that congenital vulnerability and host-dependent genetic heterogeneity are involved in the long-term effects of the infection (76, 77). Some studies have linked severe early RSV infection with allergic sensitization leading to asthma (127). Table 1 presents a list of previously conducted studies showing the increased risk of recurrent wheezing, bronchial hyperreactivity, or asthma in children after RSV-induced bronchiolitis in infancy.

TABLE 1.

Clinical and epidemiologic studies linking RSV infection to asthma

Researchers and yr (reference no.) Location No. of human subjects No. of yrs after admission to hospital with bronchiolitis Outcome/comment
Rooney and Williams, 1971 (115) Australia 62 (2-7 yrs of age) A significant association of asthma in 56% of children who subsequently experience wheeze
Gurwitz et al., 1981 (46) Canada 48 9-10 Incidence of bronchial hyperreactivity is 57%
Hall et al., 1984 (49) United States 29 8 An association between RSV infection and chronic abnormalities of pulmonary function
Mok and Simpson, 1984 (85, 86) United Kingdom 200 7 Atopy and bronchial hyperreactivity independently contribute to augmented response to RSV postinfection
Welliver and Duffy, 1993 (141) United States 43 7 Decreased pulmonary function following bronchiolitis is related to atopy
Sigurs et al., 1995 (126) Sweden 47 2 RSV infection during first yr is an important risk for asthma and allergy in the subsequent 2 yrs, especially in genetically predisposed children
Stein et al., 1999 (131) United States >180 13 Lower respiratory tract RSV infections are associated with increased risk of frequent wheeze by age 6; risk decreased markedly with age and was not significant by age 13
Sigurs et al., 2000 (125) Sweden 47 7 RSV-induced bronchiolitis severe enough to cause hospitalization is highly associated with the development of asthma and allergic sensitization at age 7.5 yrs

In a population-based birth cohort study conducted by Henderson et al. (58), a total of 150 infants (1.1% of the cohort) were admitted to the hospital within 12 months of birth with RSV-induced bronchiolitis. The prevalences of wheezing were 28.1% in the RSV group and 13.1% in the controls at 30 to 42 months and 22.6% in the RSV group and 9.6% in the controls at 69 to 81 months. The cumulative prevalences of asthma were 38.4% in the RSV group and 20.1% in controls at 91 months (58). After a study conducted in Norway, the researchers concluded that children hospitalized for early-life bronchiolitis are susceptible to recurrent wheezing and reduced pulmonary function by 7 years of age compared to age-matched children not hospitalized for early-life bronchiolitis (32). In a different study, conducted by Schauer et al. (118), a positive test for immunoglobulin E (IgE) antibodies was noted in 33% of RSV-infected children as opposed to 2.3% of children in control group. They concluded that severe RSV-induced bronchiolitis during the first year of life is an important risk factor for the development of wheezing and sensitization to common allergens during the subsequent year (118). Sigurs et al. (127) studied the outcome for 13-year-olds (46 children with RSV and 92 control subjects). They reported 43% prevalence of asthma and recurrent wheezing and 39% prevalence of allergic rhinoconjunctivitis among the RSV-infected group as opposed to 8% and 15% prevalences among the control subjects, respectively (127). Overall, the epidemiological studies have shown that RSV infection is a significant risk factor for future development of asthma, particularly in individuals who have genetic predisposition for allergic disease.

CLINICAL STUDIES

Two very important and pertinent prospective studies in relation to the link between RSV and asthma are noteworthy. The first study included 47 previously healthy infants hospitalized for RSV-induced bronchiolitis during their first year of life and a reference population of 93 infants, matched for age, sex, family history of reactive airway disease (RAD) or atopy, and general living environment and with no history of RSV infection (124). The study design included RAD, wheezing, and recurrent wheezing as the outcome measures. The allergic status of each patient was determined by skin prick testing and titers of serum IgE antibodies. By 7 years of age, 30% of children in the RSV-infected group (versus only 3% of children in the reference group) had experienced RAD. The cumulative prevalence of recurrent wheezing among children in the RSV group was twice that observed for the reference group (68% versus 34%, respectively), and the presence of any wheezing at age 7 years was 38% in the RSV-infected group and 2% in the control group. RSV-induced bronchiolitis was the only significant risk factor for RAD, whereas RSV-induced bronchiolitis, family history of atopy, and male sex were all risk factors for the presence of any wheezing. Multivariate analysis showed that the highest frequency of RAD occurred when both RSV-induced bronchiolitis and a family history of atopy were present as risk factors. That study also found a link between RSV-induced bronchiolitis and atopy; this has not been observed in other studies, possibly due to differences in the severity of RSV-induced disease and/or genetic background among the populations examined.

A second, questionnaire-based study included 519 infants enrolled in the Tucson Children's Respiratory Study who developed mild to severe RSV-induced disease without necessarily requiring hospitalization (131). On each of four occasions, when the children in the study were 6, 8, 11, and 13 years old, their parents were asked to complete a questionnaire about current parental smoking status and the child's history of wheeze. The study prospectively enrolled 1,246 infants born from 1980 to 1984. In this study, lower respiratory tract infection with RSV was associated with increased risks of both infrequent and frequent wheezing (odds ratios of 3.2 and 4.3, respectively) at age 6, but this risk was reduced as children became older, such that it was not significant at age 13. These investigators did not find a relationship between lower respiratory tract RSV infection and skin test reactivity to aeroallergens (131).

CELLULAR AND MOLECULAR LINKS

Epithelial cells are the first line of defense of the airway, and their exposure to either RSV or allergens causes signals the release of several chemokines and cytokines. These inflammatory mediators damage the epithelium and cause loss of epithelial cells, leading to increased airway permeability, mucous plugging, and decreased clearance. The degree of epithelial damage is correlated with the magnitude of inflammation and AHR (47, 54, 79, 105). Epithelial cells also regulate inflammatory events by secreting various cytokines that communicate in a paracrine manner. One of the earliest events in response to RSV infection is the initial innate cytokine response, with a time-dependent increase in expression (21). The early cytokines, including the type I interferons (IFNs), IFN-α and IFN-β, interleukin-12 (IL-12), and IL-18, have been detected in the respiratory secretions of infants with RSV infection (35, 130). Recent studies have shown that in the response to RSV infection, the airway epithelium also produces chemokines that modulate the influx of inflammatory cells into the infected tissues. These include CC (RANTES [regulated on activation, normal T-cell expressed and secreted], MCP-1 [monocyte chemoattractant protein], MIP1α [macrophage inflammatory protein 1α], and MIP1β), CXC (growth-regulated oncogene alpha [Gro-α], Gro-β, Gro-γ, IL-8, and interferon-inducible T-cell alpha chemoattractant), and CX3C (fractalkine) subclasses of chemokines in the lower airway epithelial cells (135, 144). Cytokines produced by T cells in the adaptive immune response are thought to play a role in RSV-induced disease pathogenesis. T-helper type 1 (Th1) cells stimulate cell-mediated immunity and produce inflammatory cytokines, such as IFN-γ and tumor necrosis factor alpha (TNF-α). Th2 cells stimulate humoral immune responses and produce cytokines such as IL-4, IL-5, IL-13, and IL-10 (22).

As shown in Fig. 1, RSV infection upregulates the expression of several cytokines and chemokines, such as IL-1β, IL-6, IL-8, TNF-α, MIP1α, RANTES, and the adhesion molecule ICAM-1, in cultured epithelial cells (1, 2, 25, 134). During the acute stage of infection, airway epithelial cells can initiate primary local inflammatory responses by directly responding with cytokine secretion to inflammatory stimuli or by amplifying an inflammatory event previously initiated by activated macrophages, eosinophils, or lymphocytes (Table 2) (42, 54, 63). These cell types may have important roles in controlling infection; however, inappropriate expression of inflammatory mediators may be linked to hyperresponsiveness and exacerbation of RSV-associated asthma (24, 120). Acute respiratory viral infections are often accompanied by neutrophilia of upper and lower respiratory secretions, and it is likely that products of neutrophil activation are involved in airway obstruction and lower airway symptoms (37, 81).

FIG. 1.

FIG. 1.

Epithelial cell as the target of RSV and possible role of NF-κB (hence the question mark) in mediating RSV-induced activation of genes. RSV infection upregulates the expression of several proinflammatory and allergy- and asthma-related cytokines and chemokines in cultured epithelial cells (1, 2, 25, 134). GM-CSF, granulocyte-monocyte colony-stimulating factor; FcER1α, Fc fragment of alpha subunit high-affinity (I) IgE receptor; iNOS, inducible nitric oxide synthase; MUC-1, mucin-1.

TABLE 2.

Common inflammatory cells and mediators in the upper and lower airway in RSV infection

Immune response Immune mechanism(s)
Inflammatory cells Eosinophills
Mast cells
Basophills
Dendritic cells
Neutrophills
Th2 lymphocytes
Mediators Histamine
Cysteinyl leukotrienes
Prostaglandins
Kinins
Cytokines
Chemokines

Experimental findings suggest that the cellular immune response to RSV, particularly IFN-γ production, clearly influences the clinical outcome of infection. Because the immune response to viral infections appears to have some unique features in persons with asthma, including reduced generation of IFN-γ in peripheral blood cells, asthmatics may suffer more-severe lower respiratory manifestations (93). In addition to the impact on immunogenicity due to the actions of the cytokines and chemokines they produce, bronchial epithelial cells also express adhesion molecules, such as ICAM-1, and major histocompatibility complex class II molecules, which enable them to present antigens directly to T cells. Thus, the role of bronchial epithelial cells in the viral immune response goes beyond simply being a barrier. These cells can influence the extent and potency of the adaptive immune response by means of the cytokines and chemokines they secrete, as well as the major histocompatibility complex class II and ICAM-1 surface molecules. It was shown previously that RSV colocalizes with ICAM-1 on human epithelial HEp-2 cells and that a neutralizing antibody to ICAM-1 significantly reduced RSV infection and secretion of RANTES (8).

The appearance of NF-κB in the nucleus of a RSV-infected cell coincides with an increase in IL-8 gene transcription. Moreover, RSV-induced IL-8 gene transcription requires viral replication, and the inhibition of viral replication reverses RSV-induced NF-κB activation and IL-8 transcription (19, 144). It has been proposed that RSV gene products act via the sequestration of protein phosphatases and induction of kinases that affect IκB kinases, which leads to the persistent activation of NF-κB and the cellular activation of a whole battery of cytokine and chemokine genes (10). In addition to NF-κB, the transcription factors CCAAT/enhancer-binding protein (18) and activator protein (AP-1) (70) have also been implicated in postviral replication gene activation events, including expression of the genes encoding ICAM-1 and various cytokines (70). In addition, there is increasing evidence that NF-κB is involved in chronic inflammatory diseases such as asthma (31).

The involvement of STAT proteins in the regulation of events underlying RSV infection has been implied but not demonstrated. RSV infection can upregulate the expression of IL-6, IL-8, MCP-1, TNF-α, MIP1α, RANTES, IFNs (2, 25, 134, 136), and ICAM-1, and this is partially controlled by STAT transcription factors (8). RSV increased expression of IRF1 (interferon-regulatory factor 1), ICAM-1, and RANTES in normal fibroblasts but not in those from STAT1-deficient mice, which indicated a requirement for STAT1 (70). Also, the synergy between IFN-γ and TNF-α in the transcriptional activation of genes such as those encoding IRF1 and ICAM-1 may be mediated by the cooperation of STAT1 and NF-κB (23, 84). IFN-γ, which acts as an antiviral agent, is a potent inducer of the STAT pathway (74). RSV infection activates STAT1 and STAT3 in the human epithelial carcinoma line A549 and normal human bronchial epithelial cells; however, little is known about the roles of STAT-mediated pathways during RSV infection, especially during early periods prior to RSV replication within host cells (70).

Other pathways are also affected by RSV. During the early events of RSV infection in normal human bronchial epithelial cells, the phosphokinase C-α isozyme translocates to the cell membrane and colocalizes with the viral particles adsorbed to the membrane. Inhibition of phosphokinase C-α prevents viral entry without modifying the adsorption step (116). It has been reported that RSV infection of A549 cells activates the extracellular signal-regulated kinase-1 (ERK-1) and ERK-2 pathways and that inhibition of these ERK pathways significantly decreases RSV infection of these cells compared to the case for controls (69). Blocking ICAM-1 also reduces RSV infection in A549 cells (8).

NEUROLOGICAL LINKS

An animal model for RSV-induced bronchiolitis was developed with Fischer 344 rats (105). These rats build up a strong immune response and are thus able to clear RSV from the lungs in a few weeks. In one of the studies, weanling rats were inoculated with RSV, and the long-term effects of early infection on the nonadrenergic noncholinergic nervous system (NANC) were examined (68). It was found that NANC nerves at the infection site released substance P and other peptide neurotransmitters which play an important role in the initial inflammatory process and in the modulation of the immune response to the virus. Neurogenic inflammatory responses across the respiratory tract were observed as the rats grew from infancy to adulthood.

Recent evidence suggests that RSV infection induces the expression of NK1 neurokinin receptors on T lymphocytes within bronchiole-associated lymphoid tissue (100, 102, 103). These G protein-coupled receptors respond to substance P with activation of the phosphoinositide pathway and production of inositol triphosphate. The authors hypothesized that, following virus infection, NK1 receptor-bearing lymphocytes are deployed into the airways and that subsequent neuropeptide stimulation might be involved in the release of proinflammatory cytokines.

Acute RSV infection is also associated with markedly increased numbers of mast cells in the airway mucosa, which might be involved in the observed massive expression of 5-lipoxygenase, with transient production of cysteinyl leukotrienes (138). It has been suggested that leukotrienes released as a result of the mast cell-nerve interactions during RSV infection could eventually potentiate the inflammatory effects of neuropeptides such as substance P. Based on this model, it was concluded that activation of the NANC by irritants could be responsible for the recurring airway inflammation which continues after the acute RSV infection has been cleared (103). In the most recent studies, it was shown that RSV infection promotes a large increase in the expression of nerve growth factor and neurotrophin receptors (101). Release of nerve growth factor leads to short- and long-term changes in the distribution and reactivity of sensory nerves across the respiratory tract. These may participate in the exaggerated inflammatory response seen during and after infection. It was postulated that changes in neurotrophin expression in the respiratory tract may represent an important factor in the association between susceptibility to childhood asthma and RSV-induced bronchiolitis in infancy.

RSV-ASTHMA LINK IN EXPERIMENTAL ANIMAL MODELS

Knowledge of the clinical pathogenesis of RSV-induced disease is limited because of the relatively small number of human studies. Animal models of RSV infection utilizing mice, rats, guinea pigs, sheep, cows, and monkeys have been developed to study the disease mechanism in detail, and this has resulted in the realization that RSV causes a multifaceted disease whose clinical manifestations and sequelae depend upon age, genetic makeup, immunologic status, and concurrent disease within the subpopulations. Mice have been extensively used to study the immune mechanisms of RSV-induced disease (97). However, there are significant limitations involved in using murine systems, including a lack of infection in bronchiolar epithelium, failure to spread infection from upper to lower airway, and relatively large amount of inoculum required for initiation of pathology and illness.

In BALB/c mice, a large RSV inoculum (107 PFU/ml) administered intranasally induces pneumonia, clinical illness, such as weight loss or ruffled hair, and appreciable pathology in the lung, along with infection of the lung polymorphonuclear cells and macrophages (42, 43, 63, 92). Nevertheless, many elements of the mouse and human responses to RSV infection are similar, particularly in the production of cytokines and chemokines and patterns of lung inflammation (83). The immunohistopathology of RSV infection in mice is well characterized (39-43, 63, 92), and it resembles that of human RSV infection (47). Similar to the case with humans, mice rendered immunocompromised by treatment with cyclophosphamide have increased susceptibility to RSV infection and produce higher viral titers in the lung tissue than untreated mice (132). A study to establish the importance of IFN-γ produced during the primary infection in the protection against the development of airway hyperresponsiveness on reinfection was recently conducted. For this study, both wild-type and IFN-γ-knockout mice were infected with RSV as neonates or weanlings and reinfected 5 weeks later. Airway responses were assessed on day 6 after primary or secondary infection (73). The mouse model has several advantages over models of other species. There is a vast array of inbred, congenic, transgenic, and knockout strains of mice available, and they are cheap to purchase and maintain.

The cotton rat is 100-fold more permissive (per input dose of virus) as well as more responsive immunologically than the mouse (43, 110). Studies with the cotton rat model showed that RSV-neutralizing serum IgG could prevent pulmonary infection and attenuate nasal infection, and these studies established the rationale for clinical trials of RSV prophylaxis with plasma-derived IgG for high-risk infants (44, 106, 108). Cotton rats also develop a vaccine-enhanced disease similar to that in humans and other primates (107, 109), but the lack of congenic, transgenic, or knockout strains has limited the use of these animals. Thus, most RSV studies will continue to be done with the mouse.

Although chimpanzees have the closest genetic relatedness to humans, practical and biological considerations severely limit the use of chimpanzees in RSV research. Chimpanzees are scarce, extremely expensive, and available only through primate breeding programs. Experimental RSV infections in owl monkeys, rhesus monkeys, African green monkeys, cebus monkeys, squirrel monkeys, bonnet monkeys, and baboons have also been described (9, 64, 111-113, 128). The cost and required maintenance of these species are less than those for chimpanzees, but these species still require specialized housing and handling methods.

Results with animal models have been disappointing in defining mechanisms by which RSV might lead to asthma exacerbations, largely because of the lack of reproducible results among different research groups. For instance, one group found that primary RSV infection in BALB/c mice caused IL-13-mediated increases in airway mucus and airway responsiveness to methacholine compared to the case in sham-challenged mice (133). However, this group found no airway eosinophilia in cases of RSV infection. Another group reported that RSV induced AHR and lung eosinophilia, both of which were mediated by IL-5 in BALB/c mice and not IL-13 in C57BL/6 mice (94, 119). Still another group found that primary RSV infection did not induce AHR, detectable levels of IL-5 or IL-13 mRNA or protein expression, or lung eosinophilia. However, this group found that RSV infection during allergic lung inflammation induced significant AHR and prolonged mucus production (55, 98, 99). AHR in BALB/c mice seems to be linked to both IFN-γ and leukotriene generation (140). It is important to keep in mind the relative strengths and weaknesses of the various animal models in order to better understand the mechanism by which RSV infection predisposes to asthma or exacerbates existing respiratory disease.

PROPHYLAXIS

Despite the considerable impact of RSV on respiratory health during childhood, the treatment options available for the management of lower respiratory tract RSV infections are limited and remain primarily supportive and symptomatic. While bronchodilators are used frequently for infants with acute bronchiolitis, their efficacy remains highly controversial (65, 66, 91). There have been many attempts to produce an effective RSV vaccine or anti-RSV drug but with only limited success. Tests of the first developed vaccine on human children ended with disastrous results when natural RSV infection developed in those children (up to 80% of the children were hospitalized, and two children died) (67).

Passive immunoprophylaxis is currently the only option for avoiding RSV-induced disease. The IMpact-RSV trial was a randomized, double-blind, placebo-controlled trial conducted at 139 sites in the United States, the United Kingdom, and Canada (60). It evaluated prophylaxis with palivizumab, a humanized monoclonal antibody against the RSV fusion protein, in 1,502 prematurely born children with or without chronic lung disease (CLD) by using reduction in hospitalizations for RSV as the primary datum of this study. Children received an intramuscular injection of either palivizumab (15 mg/kg of body weight) or placebo every month for 5 months. An overall 55% reduction in hospitalizations for RSV (10.6% in the placebo group hospitalized versus 4.8% in the palivizumab group hospitalized) was observed. Specifically, prematurely born children without CLD had a 78% reduction in RSV-related hospitalizations (8.1% versus 1.8%) compared with a 39% hospitalization reduction for children with CLD (12.8% versus 7.9%). A cohort of preterm infants, including 191 who had received palivizumab and were not hospitalized for RSV and 230 who had never received palivizumab, out of which 76 were hospitalized for RSV, were assessed for recurrent wheezing beginning at a mean age of 19 months and continuing for 24 months. The incidence of recurrent wheezing in the palivizumab-treated subjects (13%) was significantly lower than that in the group of untreated subjects (26%) (129). The high cost of palivizumab is the main factor for its restricted use. The cost-effectiveness of the use of palivizumab in children at high risk of hospitalization, preterm infants (≤35 weeks gestation), children with bronchopulmonary dysplasia, and children with congenital heart disease was assessed. The United Kingdom National Health Sciences study showed that palivizumab prophylaxis against severe RSV infection in children at high risk is effective in preventing hospitalization and may be cost-effective in comparison to no prophylaxis (90).

These results coupled with epidemiologic evidence of the link between RSV and RAD raise the issue of whether immunoprophylaxis against RSV can reduce the risk for development of RAD. In the Fischer 344 rat model, prophylaxis with palivizumab prevented the development of acute neurogenic inflammatory changes in the lower respiratory tract following inoculation of RSV (104). Palivizumab prophylaxis also appeared to guard against development of long-term vulnerability to neurogenically mediated inflammation.

Another potential area of anti-RSV research has been the investigation of small interfering RNAs (siRNAs) against viral proteins as a means of blocking virus replication (6). Prophylactic intranasal administration of an siRNA formulation specific for RSV P protein is able to significantly reduce the viral load and the disease parameters in RSV-infected BALB/c mice (11). Although intranasal administration of naked siRNA to humans was found to be safe in a phase I study, other studies have shown toxicity for this method.

siRNA specific for the RSV NS1 mRNA also elicited antiviral effects in BALB/c mice (143). Silencing of the NS1 gene attenuated RSV replication and boosted the immune response through increased IFN-γ production (143). The use of silenced-NS1 (siNS1) prophylaxis may be an effective method for preventing RSV-induced bronchiolitis and potentially reducing the later development of asthma associated with severe respiratory infections. The nonstructural proteins of RSV, NS1 and NS2, afford promising targets for siRNA therapy since they are produced early in the infection cycle and are necessary for survival of virus.

In a study conducted with Fischer 344 rats, prophylaxis with siNS1 significantly reduced lung RSV titers and AHR to methacholine challenge in comparison to its effect on the control group (71). Treatment of rats with siNS1 prior to RSV exposure was effective in reducing virus titers in the lungs and in preventing the inflammation and airway hyperresponsiveness associated with the infection that have been linked to development of asthma (71). A phase I study using nanoparticle-incorporated siNS1 is currently under development, and it may have future implications in prophylaxis/therapy globally.

CONCLUSION

Despite progress in the treatment of asthma, the incidence of asthma appears to be increasing in all age groups, including children. RSV-induced bronchiolitis constitutes a proven risk factor for the development of childhood asthma. The evidence from epidemiologic and clinical studies has been considered in the debate about the role of RSV-induced bronchiolitis in childhood asthma. Some of the variations between results have come from the study design and the geographic regions and the population of subjects being studied. Cellular and molecular studies of RSV infection in human cell lines and neurologic evidence in animal models have demonstrated a possible causal relationship between RSV-induced bronchiolitis during infancy and asthma later in life. Prophylaxis with antibodies or siRNAs in animal models has been shown to prevent acute infections and offer long-term protection from chronic lung diseases. Nonetheless, further studies with humans, particularly children, are needed to verify this. Such studies are important, as they are expected to demonstrate whether achieving successful prophylaxis against RSV infection might reduce the risk for development of asthma in children later in life.

Acknowledgments

This work was supported by VA Career Scientist and Merit Review awards and NIH grant RO1 HL 71101A1 to S.S.M. and by the Joy McCann Culverhouse and Mabel and Ellsworth Simmons endowments to the University of South Florida Division of Allergy and Immunology Airway Disease Research Center.

We acknowledge the assistance of Gary B. Hellerman for critical reading and editing of the manuscript.

REFERENCES

  • 1.Arnold, R., B. Humbert, H. Werchau, H. Gallati, and W. Konig. 1994. Interleukin-8, interleukin-6, and soluble tumor necrosis factor receptor type I release from a human pulmonary epithelial-cell line (A549) exposed to respiratory syncytial virus. Immunology 82:126-133. [PMC free article] [PubMed] [Google Scholar]
  • 2.Arnold, R., F. Werner, B. Humbert, H. Werchau, and W. Konig. 1994. Effect of respiratory syncytial virus-antibody complexes on cytokine (Il-8, Il-6, Tnf-alpha) release and respiratory burst in human granulocytes. Immunology 82:184-191. [PMC free article] [PubMed] [Google Scholar]
  • 3.Arslańagic, E., M. Matsumoto, K. Suzuki, K. Nerome, H. Tsutsumi, and T. Hung. 1996. Maturation of respiratory syncytial virus within HEp-2 cell cytoplasm. Acta Virol. 40:209-214. [PubMed] [Google Scholar]
  • 4.Atreya, P. L., and S. Kulkarni. 1999. Respiratory syncytial virus strain A2 is resistant to the antiviral effects of type I interferons and human MxA. Virology 261:227-241. [DOI] [PubMed] [Google Scholar]
  • 5.Bächi, T., and C. Howe. 1973. Morphogenesis and ultrastructure of respiratory syncytial virus. J. Virol. 12:1173-1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Barik, S. 2004. Control of nonsegmented negative-strand RNA virus replication by siRNA. Virus Res. 102:27-35. [DOI] [PubMed] [Google Scholar]
  • 7.Becker, S., W. Reed, F. W. Henderson, and T. L. Noah. 1997. RSV infection of human airway epithelial cells causes production of the beta-chemokine RANTES. Am. J. Physiol. 272:L512-L520. [DOI] [PubMed] [Google Scholar]
  • 8.Behera, A. K., H. Matsuse, M. Kumar, X. Kong, R. F. Lockey, and S. S. Mohapatra. 2001. Blocking intercellular adhesion molecule-1 on human epithelial cells decreases respiratory syncytial virus infection. Biochem. Biophys. Res. Commun. 280:188-195. [DOI] [PubMed] [Google Scholar]
  • 9.Belshe, R. B., L. S. Richardson, W. T. London, D. L. Sly, J. H. Lorfeld, E. Camargo, D. A. Prevar, and R. M. Chanock. 1977. Experimental respiratory syncytial virus infection of four species of primates. J. Med. Virol. 1:157-162. [DOI] [PubMed] [Google Scholar]
  • 10.Bitko, V., and S. Barik. 1998. Persistent activation of RelA by respiratory syncytial virus involves protein kinase C, underphosphorylated IκBβ, and sequestration of protein phosphatase 2A by the viral phosphoprotein. J. Virol. 72:5610-5618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bitko, V., A. Musiyenko, O. Shulyayeva, and S. Barik. 2005. Inhibition of respiratory viruses by nasally administered siRNA. Nat. Med. 11:50-55. [DOI] [PubMed] [Google Scholar]
  • 12.Brandenburg, A. H., H. J. Neijens, and A. Osterhaus. 2001. Pathogenesis of RSV lower respiratory tract infection: implications for vaccine development. Vaccine 19:2769-2782. [DOI] [PubMed] [Google Scholar]
  • 13.Brown, G., J. Aitken, H. W. Rixon, and R. J. Sugrue. 2002. Caveolin-1 is incorporated into mature respiratory syncytial virus particles during virus assembly on the surface of virus-infected cells. J. Gen. Virol. 83:611-621. [DOI] [PubMed] [Google Scholar]
  • 14.Brown, G., C. E. Jeffree, T. McDonald, H. W. Rixon, J. D. Aitken, and R. J. Sugrue. 2004. Analysis of the interaction between respiratory syncytial virus and lipid-rafts in Hep2 cells during infection. Virology 327:175-185. [DOI] [PubMed] [Google Scholar]
  • 15.Brown, G., H. W. Rixon, and R. J. Sugrue. 2002. Respiratory syncytial virus assembly occurs in GM1-rich regions of the host-cell membrane and alters the cellular distribution of tyrosine phosphorylated caveolin-1. J. Gen. Virol. 83:1841-1850. [DOI] [PubMed] [Google Scholar]
  • 16.Centers for Disease Control and Prevention. 2006. Respiratory syncitial virus activity—United States, 2005-2006. MMWR Morb. Mortal. Wkly. Rep. 55:1277-1279. [PubMed] [Google Scholar]
  • 17.Centers for Disease Control and Prevention. 2007. Respiratory syncitial virus activity—United States, July 2006-November 2007. MMWR Morb. Mortal. Wkly. Rep. 56:1263-1265. [PubMed] [Google Scholar]
  • 18.Chini, B. A., M. A. Fiedler, L. Milligan, T. Hopkins, and J. M. Stark. 1998. Essential roles of NF-κB and C/EBP in the regulation of intercellular adhesion molecule-1 after respiratory syncytial virus infection of human respiratory epithelial cell cultures. J. Virol. 72:1623-1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Choudhary, S., S. Boldogh, R. Garofalo, M. Jamaluddin, and A. R. Brasier. 2005. Respiratory syncytial virus influences NF-κB-dependent gene expression through a novel pathway involving MAP3K14/NIK expression and nuclear complex formation with NF-κB2. J. Virol. 79:8948-8959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Collins, P. L., M. G. Hill, J. Cristina, and H. Grosfeld. 1996. Transcription elongation factor of respiratory syncytial virus, a nonsegmented negative-strand RNA virus. Proc. Natl. Acad. Sci. USA 93:81-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Collins, P. L., K. McIntosh, and R. M. Chanock. 1996. Respiratory syncytial virus, p. 1313-1351. In B. N. Fields, D. M. Knipe, P. M. Howley, et al. (ed.), Fields virology, 3rd ed. Lippincott-Raven Publishers, New York, NY.
  • 22.Constant, S. L., and K. Bottomly. 1997. Induction of Th1 and Th2 CD4+ T cell responses: the alternative approaches. Annu. Rev. Immunol. 15:297-322. [DOI] [PubMed] [Google Scholar]
  • 23.Doly, J., A. Civas, S. Navarro, and G. Uze. 1998. Type I interferons: expression and signalization. Cell. Mol. Life Sci. 54:1109-1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ehlenfield, D. R., K. Cameron, and R. C. Welliver. 2000. Eosinophilia at the time of respiratory syncytial virus bronchiolitis predicts childhood reactive airway disease. Pediatrics 105:79-83. [DOI] [PubMed] [Google Scholar]
  • 25.Elias, J. A., T. Zheng, O. Einarsson, M. Landry, T. Trow, N. Rebert, and J. Panuska. 1994. Epithelial interleukin-11. Regulation by cytokines, respiratory syncytial virus, and retinoic acids. J. Biol. Chem. 269:22261-22268. [PubMed] [Google Scholar]
  • 26.Evans, J. E., P. A. Cane, and C. R. Pringle. 1996. Expression and characterisation of the NS1 and NS2 proteins of respiratory syncytial virus. Virus Res. 43:155-161. [DOI] [PubMed] [Google Scholar]
  • 27.Falsey, A. R., P. A. Hennessey, M. A. Formica, C. Cox, and E. E. Walsh. 2005. Respiratory syncytial virus infection in elderly and high-risk adults. N. Engl. J. Med. 352:1749-1759. [DOI] [PubMed] [Google Scholar]
  • 28.Fearns, R., and P. L. Collins. 1999. Role of the M2-1 transcription antitermination protein of respiratory syncytial virus in sequential transcription. J. Virol. 73:5852-5864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Feldman, S. A., R. L. Crim, S. A. Audet, and J. A. Beeler. 2001. Human respiratory syncytial virus surface glycoproteins F, G and SH form an oligomeric complex. Arch. Virol. 146:2369-2383. [DOI] [PubMed] [Google Scholar]
  • 30.Ferris, J. A., W. A. Aherne, W. S. Locke, J. McQuillin, and P. S. Gardner. 1973. Sudden and unexpected deaths in infants: histology and virology. Br. Med. J. 2:439-442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fiedler, M. A., K. Wernke-Dollries, and J. M. Stark. 1995. Respiratory syncytial virus increases IL-8 gene expression and protein release in A549 cells. Am. J. Physiol. 269:L865-L872. [DOI] [PubMed] [Google Scholar]
  • 32.Fjaerli, H. O., T. Farstad, G. Rod, G. K. Ufert, P. Gulbrandsen, and B. Nakstad. 2005. Acute bronchiolitis in infancy as risk factor for wheezing and reduced pulmonary function by seven years in Akershus County, Norway. BMC Pediatr. 5:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Garofalo, R., F. Mei, R. Espejo, G. Ye, H. Haeberle, S. Baron, P. L. Ogra, and V. E. Reyes. 1996. Respiratory syncytial virus infection of human respiratory epithelial cells up-regulates class I MHC expression through the induction of IFN-beta and IL-1 alpha. J. Immunol. 157:2506-2513. [PubMed] [Google Scholar]
  • 34.Reference deleted.
  • 35.Garofalo, R. P., K. H. Hintz, V. Hill, P. L. Ogra, and R. C. Welliver, Sr. 2004. Production of interferon gamma in respiratory syncytial virus infection of humans is not associated with interleukins 12 and 18. J. Med. Virol. 73:289-294. [DOI] [PubMed] [Google Scholar]
  • 36.Gazala, E., V. Ron-Feldman, M. Alterman, S. Kama, and L. Novack. 2006. The association between birth season and future development of childhood asthma. Pediatr. Pulmonol. 41:1125-1128. [DOI] [PubMed] [Google Scholar]
  • 37.Gern, J. E., and W. W. Busse. 2002. Relationship of viral infections to wheezing illnesses and asthma. Nat. Rev. Immunol. 2:132-138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Glezen, W. P., L. H. Taber, A. L. Frank, and J. A. Kasel. 1986. Risk of primary infection and reinfection with respiratory syncytial virus. Am. J. Dis. Child. 140:543-546. [DOI] [PubMed] [Google Scholar]
  • 39.Graham, B. S. 1996. Immunological determinants of disease caused by respiratory syncytial virus. Trends Microbiol. 4:290-294. [DOI] [PubMed] [Google Scholar]
  • 40.Graham, B. S., L. A. Bunton, P. F. Wright, and D. T. Karzon. 1991. Role of lymphocyte-T subsets in the pathogenesis of primary infection and rechallenge with respiratory syncytial virus in mice. J. Clin. Investig. 88:1026-1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Graham, B. S., G. S. Henderson, Y. W. Tang, X. T. Lu, K. M. Neuzil, and D. G. Colley. 1993. Priming immunization determines T-helper cytokine messenger-RNA expression patterns in lungs of mice challenged with respiratory syncytial virus. J. Immunol. 151:2032-2040. [PubMed] [Google Scholar]
  • 42.Graham, B. S., T. R. Johnson, and R. S. Peebles. 2000. Immune-mediated disease pathogenesis in respiratory syncytial virus infection. Immunopharmacology 48:237-247. [DOI] [PubMed] [Google Scholar]
  • 43.Graham, B. S., M. D. Perkins, P. F. Wright, and D. T. Karzon. 1988. Primary respiratory syncytial virus-infection in mice. J. Med. Virol. 26:153-162. [DOI] [PubMed] [Google Scholar]
  • 44.Groothuis, J. R., E. A. Simoes, M. J. Levin, C. B. Hall, C. E. Long, W. J. Rodriguez, J. Arrobio, H. C. Meissner, D. R. Fulton, R. C. Welliver, et al. 1993. Prophylactic administration of respiratory syncytial virus immune globulin to high-risk infants and young children. N. Engl. J. Med. 329:1524-1530. [DOI] [PubMed] [Google Scholar]
  • 45.Grosfeld, H., M. G. Hill, and P. L. Collins. 1995. RNA replication by respiratory syncytial virus (RSV) is directed by the N, P, and L proteins; transcription also occurs under these conditions but requires RSV superinfection for efficient synthesis of full-length mRNA. J. Virol. 69:5677-5686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gurwitz, D., C. Mindorff, and H. Levison. 1981. Increased incidence of bronchial reactivity in children with a history of bronchiolitis. J. Pediatr. 98:551-555. [DOI] [PubMed] [Google Scholar]
  • 47.Hacking, D., and J. Hull. 2002. Respiratory syncytial virus—viral biology and the host response. J. Infect. 45:18-24. [DOI] [PubMed] [Google Scholar]
  • 48.Haeberle, H. A., A. Casola, Z. Gatalica, S. Petronella, H. J. Dieterich, P. B. Ernst, A. R. Brasier, and R. P. Garofalo. 2004. IκB kinase is a critical regulator of chemokine expression and lung inflammation in respiratory syncytial virus infection. J. Virol. 78:2232-2241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hall, C. B., W. J. Hall, C. L. Gala, F. B. MaGill, and J. P. Leddy. 1984. Long-term prospective study in children after respiratory syncytial virus infection. J. Pediatr. 105:358-364. [DOI] [PubMed] [Google Scholar]
  • 50.Hall, C. B., W. J. Hall, and D. M. Speers. 1979. Clinical and physiological manifestations of bronchiolitis and pneumonia. Outcome of respiratory syncytial virus. Am. J. Dis. Child. 133:798-802. [DOI] [PubMed] [Google Scholar]
  • 51.Hall, C. B., C. E. Long, and K. C. Schnabel. 2001. Respiratory syncytial virus infections in previously healthy working adults. Clin. Infect. Dis. 33:792-796. [DOI] [PubMed] [Google Scholar]
  • 52.Hallak, L. K., P. L. Collins, W. Knudson, and M. E. Peeples. 2000. Iduronic acid-containing glycosaminoglycans on target cells are required for efficient respiratory syncytial virus infection. Virology 271:264-275. [DOI] [PubMed] [Google Scholar]
  • 53.Hardy, R. W., and G. W. Wertz. 1998. The product of the respiratory syncytial virus M2 gene ORF1 enhances readthrough of intergenic junctions during viral transcription. J. Virol. 72:520-526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Harris, J., and D. Werling. 2003. Binding and entry of respiratory syncytial virus into host cells and initiation of the innate immune response. Cell. Microbiol. 5:671-680. [DOI] [PubMed] [Google Scholar]
  • 55.Hashimoto, K., B. S. Graham, S. B. Ho, K. B. Adler, R. D. Collins, S. J. Olson, W. Zhou, T. Suzutani, P. W. Jones, K. Goleniewska, J. F. O'Neal, and R. S. Peebles, Jr. 2004. Respiratory syncytial virus in allergic lung inflammation increases Muc5ac and gob-5. Am. J. Respir. Crit. Care Med. 170:306-312. [DOI] [PubMed] [Google Scholar]
  • 56.Henderson, F. W., W. A. Clyde, Jr., A. M. Collier, F. W. Denny, R. J. Senior, C. I. Sheaffer, W. G. Conley III, and R. M. Christian. 1979. The etiologic and epidemiologic spectrum of bronchiolitis in pediatric practice. J. Pediatr. 95:183-190. [DOI] [PubMed] [Google Scholar]
  • 57.Henderson, F. W., A. M. Collier, W. A. Clyde, Jr., and F. W. Denny. 1979. Respiratory-syncytial-virus infections, reinfections and immunity. A prospective, longitudinal study in young children. N. Engl. J. Med. 300:530-534. [DOI] [PubMed] [Google Scholar]
  • 58.Henderson, J., T. N. Hilliard, A. Sherriff, D. Stalker, N. Al Shammari, and H. M. Thomas. 2005. Hospitalization for RSV bronchiolitis before 12 months of age and subsequent asthma, atopy and wheeze: a longitudinal birth cohort study. Pediatr. Allergy Immunol. 16:386-392. [DOI] [PubMed] [Google Scholar]
  • 59.Huang, Y. T., P. L. Collins, and G. W. Wertz. 1985. Characterization of the 10 proteins of human respiratory syncytial virus: identification of a fourth envelope-associated protein. Virus Res. 2:157-173. [DOI] [PubMed] [Google Scholar]
  • 60.IMpact-RSV Study Group. 1998. Palivizumab, a humanized respiratory syncytial virus monoclonal antibody, reduces hospitalization from respiratory syncytial virus infection in high-risk infants. Pediatrics 102:531-537. [PubMed] [Google Scholar]
  • 61.Jamaluddin, M., R. Garofalo, P. L. Ogra, and A. R. Brasier. 1996. Inducible translational regulation of the NF-IL6 transcription factor by respiratory syncytial virus infection in pulmonary epithelial cells. J. Virol. 70:1554-1563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Jeffree, C. E., H. W. Rixon, G. Brown, J. Aitken, and R. J. Sugrue. 2003. Distribution of the attachment (G) glycoprotein and GM1 within the envelope of mature respiratory syncytial virus filaments revealed using field emission scanning electron microscopy. Virology 306:254-267. [DOI] [PubMed] [Google Scholar]
  • 63.Johnson, T. R., S. M. Hong, L. Van Kaer, Y. Koezuka, and B. S. Graham. 2002. NK T cells contribute to expansion of CD8+ T cells and amplification of antiviral immune responses to respiratory syncytial virus. J. Virol. 76:4294-4303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kakuk, T. J., K. Soike, R. J. Brideau, R. M. Zaya, S. L. Cole, J. Y. Zhang, E. D. Roberts, P. A. Wells, and M. W. Wathen. 1993. A human respiratory syncytial virus (RSV) primate model of enhanced pulmonary pathology induced with a formalin-inactivated RSV vaccine but not a recombinant FG subunit vaccine. J. Infect. Dis. 167:553-561. [DOI] [PubMed] [Google Scholar]
  • 65.Kellner, J. D., A. Ohlsson, A. M. Gadomski, and E. E. Wang. 2000. Bronchodilators for bronchiolitis. Cochrane Database Syst. Rev. 2000:CD001266. [DOI] [PubMed] [Google Scholar]
  • 66.Kellner, J. D., A. Ohlsson, A. M. Gadomski, and E. E. Wang. 1996. Efficacy of bronchodilator therapy in bronchiolitis. A meta-analysis. Arch. Pediatr. Adolesc. Med. 150:1166-1172. [DOI] [PubMed] [Google Scholar]
  • 67.Kim, H. W., J. G. Canchola, C. D. Brandt, G. Pyles, R. M. Chanock, K. Jensen, and R. H. Parrott. 1969. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am. J. Epidemiol. 89:422-434. [DOI] [PubMed] [Google Scholar]
  • 68.King, K. A., C. Hu, M. M. Rodriguez, R. Romaguera, X. Jiang, and G. Piedimonte. 2001. Exaggerated neurogenic inflammation and substance P receptor upregulation in RSV-infected weanling rats. Am. J. Respir. Cell Mol. Biol. 24:101-107. [DOI] [PubMed] [Google Scholar]
  • 69.Kong, X., H. San Juan, A. Behera, M. E. Peeples, J. Wu, R. F. Lockey, and S. S. Mohapatra. 2004. ERK-1/2 activity is required for efficient RSV infection. FEBS Lett. 559:33-38. [DOI] [PubMed] [Google Scholar]
  • 70.Kong, X., H. San Juan, M. Kumar, A. K. Behera, A. Mohapatra, G. R. Hellermann, S. Mane, R. F. Lockey, and S. S. Mohapatra. 2003. Respiratory syncytial virus infection activates STAT signaling in human epithelial cells. Biochem. Biophys. Res. Commun. 306:616-622. [DOI] [PubMed] [Google Scholar]
  • 71.Kong, X., W. Zhang, R. F. Lockey, A. Auais, G. Piedimonte, and S. S. Mohapatra. 2007. Respiratory syncytial virus infection in Fischer 344 rats is attenuated by short interfering RNA against the RSV-NS1 gene. Genet. Vaccines Ther. 5:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kurt-Jones, E. A., L. Popova, L. Kwinn, L. M. Haynes, L. P. Jones, R. A. Tripp, E. E. Walsh, M. W. Freeman, D. T. Golenbock, L. J. Anderson, and R. W. Finberg. 2000. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat. Immunol. 1:398-401. [DOI] [PubMed] [Google Scholar]
  • 73.Lee, Y. M., N. Miyahara, K. Takeda, J. Prpich, A. Oh, A. Balhorn, A. Joetham, E. W. Gelfand, and A. Dakhama. 2008. IFN-gamma production during initial infection determines the outcome of reinfection with respiratory syncytial virus. Am. J. Respir. Crit. Care Med. 177:208-218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Liu, P., M. Jamaluddin, K. Li, R. P. Garofalo, A. Casola, and A. R. Brasier. 2007. Retinoic acid-inducible gene I mediates early antiviral response and Toll-like receptor 3 expression in respiratory syncytial virus-infected airway epithelial cells. J. Virol. 81:1401-1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Malhotra, R., M. Ward, H. Bright, R. Priest, M. R. Foster, M. Hurle, E. Blair, and M. Bird. 2003. Isolation and characterisation of potential respiratory syncytial virus receptor(s) on epithelial cells. Microbes Infect. 5:123-133. [DOI] [PubMed] [Google Scholar]
  • 76.Martinez, F. D. 2005. Gene-environment interactions in asthma and allergies: a new paradigm to understand disease causation. Immunol. Allergy Clin. N. Am. 25:709-721. [DOI] [PubMed] [Google Scholar]
  • 77.Martinez, F. D., G. Antognoni, F. Macri, E. Bonci, F. Midulla, G. De Castro, and R. Ronchetti. 1988. Parental smoking enhances bronchial responsiveness in nine-year-old children. Am. Rev. Respir. Dis. 138:518-523. [DOI] [PubMed] [Google Scholar]
  • 78.Martínez, I., and J. A. Melero. 2000. Binding of human respiratory syncytial virus to cells: implication of sulfated cell surface proteoglycans. J. Gen. Virol. 81:2715-2722. [DOI] [PubMed] [Google Scholar]
  • 79.Matsuse, H., A. K. Behera, M. Kumar, H. Rabb, R. F. Lockey, and S. S. Mohapatra. 2000. Recurrent respiratory syncytial virus infections in allergen-sensitized mice lead to persistent airway inflammation and hyperresponsiveness. J. Immunology 164:6583-6592. [DOI] [PubMed] [Google Scholar]
  • 80.McCurdy, L. H., and B. S. Graham. 2003. Role of plasma membrane lipid microdomains in respiratory syncytial virus filament formation. J. Virol. 77:1747-1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.McNamara, P. S., and R. L. Smyth. 2002. The pathogenesis of respiratory syncytial virus disease in childhood. Br. Med. Bull. 61:13-28. [DOI] [PubMed] [Google Scholar]
  • 82.Mejías, A., and O. Ramilo. 2002. Asthma and respiratory syncytial virus. Myth or reality? An. Esp. Pediatr. 57:199-204. (In Spanish.) [PubMed] [Google Scholar]
  • 83.Miller, A. L., T. L. Bowlin, and N. W. Lukacs. 2004. Respiratory syncytial virus-induced chemokine production: linking viral replication to chemokine production in vitro and in vivo. J. Infect. Dis. 189:1419-1430. [DOI] [PubMed] [Google Scholar]
  • 84.Mohty, M., A. Vialle-Castellano, J. A. Nunes, D. Isnardon, D. Olive, and W. Gaugler. 2003. IFN-alpha skews monocyte differentiation into toll-like receptor 7-expressing dendritic cells with potent functional activities. J. Immunol. 171:3385-3393. [DOI] [PubMed] [Google Scholar]
  • 85.Mok, J. Y., and H. Simpson. 1984. Outcome for acute bronchitis, bronchiolitis, and pneumonia in infancy. Arch. Dis. Child. 59:306-309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Mok, J. Y., and H. Simpson. 1984. Symptoms, atopy, and bronchial reactivity after lower respiratory infection in infancy. Arch. Dis. Child. 59:299-305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mullins, J. A., A. C. Lamonte, J. S. Bresee, and L. J. Anderson. 2003. Substantial variability in community respiratory syncytial virus season timing. Pediatr. Infect. Dis. J. 22:857-862. [DOI] [PubMed] [Google Scholar]
  • 88.Noah, T. L., and S. Becker. 1993. Respiratory syncytial virus-induced cytokine production by a human bronchial epithelial cell line. Am. J. Physiol. 265:L472-L478. [DOI] [PubMed] [Google Scholar]
  • 89.Norrby, E., H. Marusyk, and C. Orvell. 1970. Ultrastructural studies of the multiplication of RS (respiratory syncytial) virus. Acta Pathol. Microbiol. Scand. B 78:268. [PubMed] [Google Scholar]
  • 90.Nuijten, M. J., W. Wittenberg, and M. Lebmeier. 2007. Cost effectiveness of palivizumab for respiratory syncytial virus prophylaxis in high-risk children: a UK analysis. Pharmacoeconomics 25:55-71. [DOI] [PubMed] [Google Scholar]
  • 91.Offer, I., S. Ashkenazi, G. Livni, and I. Shalit. 2000. The diagnostic and therapeutic approach to acute bronchiolitis in hospitalized children in Israel: a nationwide survey. Isr. Med. Assoc. J. 2:108-110. [PubMed] [Google Scholar]
  • 92.Openshaw, P. J. M. 1995. Immunity and immunopathology to respiratory syncytial virus—the mouse model. Am. J. Respir. Crit. Care Med. 152:S59-S62. [DOI] [PubMed] [Google Scholar]
  • 93.Papadopoulos, N. G., M. Moustaki, M. Tsolia, A. Bossios, E. Astra, A. Prezerakou, D. Gourgiotis, and D. Kafetzis. 2002. Association of rhinovirus infection with increased disease severity in acute bronchiolitis. Am. J. Respir. Crit. Care Med. 165:1285-1289. [DOI] [PubMed] [Google Scholar]
  • 94.Park, J. W., C. Taube, E. S. Yang, A. Joetham, A. Balhorn, K. Takeda, N. Miyahara, A. Dakhama, D. D. Donaldson, and E. W. Gelfand. 2003. Respiratory syncytial virus-induced airway hyperresponsiveness is independent of IL-13 compared with that induced by allergen. J. Allergy Clin. Immunol. 112:1078-1087. [DOI] [PubMed] [Google Scholar]
  • 95.Parrott, R. H., H. W. Kim, J. O. Arrobio, D. S. Hodes, B. R. Murphy, C. D. Brandt, E. Camargo, and R. M. Chanock. 1973. Epidemiology of respiratory syncytial virus infection in Washington, D.C. II. Infection and disease with respect to age, immunologic status, race and sex. Am. J. Epidemiol. 98:289-300. [DOI] [PubMed] [Google Scholar]
  • 96.Parry, J. E., P. V. Shirodaria, and C. R. Pringle. 1979. Pneumoviruses: the cell surface of lytically and persistently infected cells. J. Gen. Virol. 44:479-491. [DOI] [PubMed] [Google Scholar]
  • 97.Peebles, R. S., Jr., and B. S. Graham. 2005. Pathogenesis of respiratory syncytial virus infection in the murine model. Proc. Am. Thorac. Soc. 2:110-115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Peebles, R. S., Jr., J. R. Sheller, R. D. Collins, A. K. Jarzecka, D. B. Mitchell, R. A. Parker, and B. S. Graham. 2001. Respiratory syncytial virus infection does not increase allergen-induced type 2 cytokine production, yet increases airway hyperresponsiveness in mice. J. Med. Virol. 63:178-188. [PubMed] [Google Scholar]
  • 99.Peebles, R. S., Jr., J. R. Sheller, J. E. Johnson, D. B. Mitchell, and B. S. Graham. 1999. Respiratory syncytial virus infection prolongs methacholine-induced airway hyperresponsiveness in ovalbumin-sensitized mice. J. Med. Virol. 57:186-192. [DOI] [PubMed] [Google Scholar]
  • 100.Piedimonte, G. 2002. The association between respiratory syncytial virus infection and reactive airway disease. Respir. Med. 96(Suppl. B):S25-S29. [PubMed] [Google Scholar]
  • 101.Piedimonte, G. 2003. Contribution of neuroimmune mechanisms to airway inflammation and remodeling during and after respiratory syncytial virus infection. Pediatr. Infect. Dis. J. 22:S66-S75. [DOI] [PubMed] [Google Scholar]
  • 102.Piedimonte, G. 2002. Neuroimmune interactions in respiratory syncytial virus-infected airways. Pediatr. Infect. Dis. J. 21:462-467. [DOI] [PubMed] [Google Scholar]
  • 103.Piedimonte, G. 2002. Pathophysiological mechanisms for the respiratory syncytial virus-reactive airway disease link. Respir. Res. 3(Suppl. 1):S21-S25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Piedimonte, G., K. A. King, N. L. Holmgren, P. J. Bertrand, M. M. Rodriguez, and R. L. Hirsch. 2000. A humanized monoclonal antibody against respiratory syncytial virus (palivizumab) inhibits RSV-induced neurogenic-mediated inflammation in rat airways. Pediatr. Res. 47:351-356. [DOI] [PubMed] [Google Scholar]
  • 105.Piedimonte, G., M. M. Rodriguez, K. A. King, S. McLean, and X. B. Jiang. 1999. Respiratory syncytial virus upregulates expression of the substance P receptor in rat lungs. Am. J. Physiol. 277:L831-L840. [DOI] [PubMed] [Google Scholar]
  • 106.PREVENT Study Group. 1997. Reduction of respiratory syncytial virus hospitalization among premature infants and infants with bronchopulmonary dysplasia using respiratory virus immune globulin prophylaxis. Pediatrics 99:93-99. [DOI] [PubMed] [Google Scholar]
  • 107.Prince, G. A., V. G. Hemming, and R. M. Chanock. 1986. The use of purified immunoglobulin in the therapy of respiratory syncytial virus infection. Pediatr. Infect. Dis. 5:S201-S203. [PubMed] [Google Scholar]
  • 108.Prince, G. A., V. G. Hemming, R. L. Horswood, and R. M. Chanock. 1985. Immunoprophylaxis and immunotherapy of respiratory syncytial virus infection in the cotton rat. Virus Res. 3:193-206. [DOI] [PubMed] [Google Scholar]
  • 109.Prince, G. A., A. B. Jenson, V. G. Hemming, B. R. Murphy, E. E. Walsh, R. L. Horswood, and R. M. Chanock. 1986. Enhancement of respiratory syncytial virus pulmonary pathology in cotton rats by prior intramuscular inoculation of formalin-inactivated virus. J. Virol. 57:721-728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Prince, G. A., L. Potash, R. L. Horswood, E. Camargo, S. C. Suffin, R. A. Johnson, and R. M. Chanock. 1979. Intramuscular inoculation of live respiratory syncytial virus induces immunity in cotton rats. Infect. Immun. 23:723-728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Prince, G. A., S. C. Suffin, D. A. Prevar, E. Camargo, D. L. Sly, W. T. London, and R. M. Chanock. 1979. Respiratory syncytial virus infection in owl monkeys: viral shedding, immunological response, and associated illness caused by wild-type virus and two temperature-sensitive mutants. Infect. Immun. 26:1009-1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Richardson, L. S., R. B. Belshe, W. T. London, D. L. Sly, D. A. Prevar, E. Camargo, and R. M. Chanock. 1978. Evaluation of five temperature-sensitive mutants of respiratory syncytial virus in primates. I. Viral shedding, immunologic response, and associated illness. J. Med. Virol. 3:91-100. [DOI] [PubMed] [Google Scholar]
  • 113.Richardson, L. S., R. B. Belshe, D. L. Sly, W. T. London, D. A. Prevar, E. Camargo, and R. M. Chanock. 1978. Experimental respiratory syncytial virus pneumonia in cebus monkeys. J. Med. Virol. 2:45-59. [DOI] [PubMed] [Google Scholar]
  • 114.Roberts, S. R., R. W. Compans, and G. W. Wertz. 1995. Respiratory syncytial virus matures at the apical surfaces of polarized epithelial cells. J. Virol. 69:2667-2673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Rooney, J. C., and H. E. Williams. 1971. The relationship between proved viral bronchiolitis and subsequent wheezing. J. Pediatr. 79:744-747. [DOI] [PubMed] [Google Scholar]
  • 116.San-Juan-Vergara, H., M. E. Peeples, R. F. Lockey, and S. S. Mohapatra. 2004. Protein kinase C-alpha activity is required for respiratory syncytial virus fusion to human bronchial epithelial cells. J. Virol. 78:13717-13726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Schaller, M., C. M. Hogaboam, N. Lukacs, and S. L. Kunkel. 2006. Respiratory viral infections drive chemokine expression and exacerbate the asthmatic response. J. Allergy Clin. Immunol. 118:295-304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Schauer, U., S. Hoffjan, J. Bittscheidt, A. Kochling, S. Hemmis, S. Bongartz, and V. Stephan. 2002. RSV bronchiolitis and risk of wheeze and allergic sensitisation in the first year of life. Eur. Respir. J. 20:1277-1283. [DOI] [PubMed] [Google Scholar]
  • 119.Schwarze, J., G. Cieslewicz, E. Hamelmann, A. Joetham, L. D. Shultz, M. C. Lamers, and E. W. Gelfand. 1999. IL-5 and eosinophils are essential for the development of airway hyperresponsiveness following acute respiratory syncytial virus infection. J. Immunol. 162:2997-3004. [PubMed] [Google Scholar]
  • 120.Schwarze, J., E. Hamelmann, K. L. Bradley, K. Takeda, and E. W. Gelfand. 1997. Respiratory syncytial virus infection results in airway hyperresponsiveness and enhanced airway sensitization to allergen. J. Clin. Investig. 100:226-233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Schwarze, J., D. R. O'Donnell, A. Rohwedder, and P. J. Openshaw. 2004. Latency and persistence of respiratory syncytial virus despite T cell immunity. Am. J. Respir. Crit. Care Med. 169:801-805. [DOI] [PubMed] [Google Scholar]
  • 122.Shay, D. K., R. C. Holman, R. D. Newman, L. L. Liu, J. W. Stout, and L. J. Anderson. 1999. Bronchiolitis-associated hospitalizations among US children, 1980-1996. JAMA 282:1440-1446. [DOI] [PubMed] [Google Scholar]
  • 123.Shay, D. K., R. C. Holman, G. E. Roosevelt, M. J. Clarke, and L. J. Anderson. 2001. Bronchiolitis-associated mortality and estimates of respiratory syncytial virus-associated deaths among US children, 1979-1997. J. Infect. Dis. 183:16-22. [DOI] [PubMed] [Google Scholar]
  • 124.Sigurs, N. 2002. A cohort of children hospitalised with acute RSV bronchiolitis: impact on later respiratory disease. Paediatr. Respir. Rev. 3:177-183. [DOI] [PubMed] [Google Scholar]
  • 125.Sigurs, N., R. Bjarnason, F. Sigurbergsson, and B. Kjellman. 2000. Respiratory syncytial virus bronchiolitis in infancy is an important risk factor for asthma and allergy at age 7. Am. J. Respir. Crit. Care Med. 161:1501-1507. [DOI] [PubMed] [Google Scholar]
  • 126.Sigurs, N., R. Bjarnason, F. Sigurbergsson, B. Kjellman, and B. Bjorksten. 1995. Asthma and immunoglobulin E antibodies after respiratory syncytial virus bronchiolitis: a prospective cohort study with matched controls. Pediatrics 95:500-505. [PubMed] [Google Scholar]
  • 127.Sigurs, N., P. M. Gustafsson, R. Bjarnason, F. Lundberg, S. Schmidt, F. Sigurbergsson, and B. Kjellman. 2005. Severe respiratory syncytial virus bronchiolitis in infancy and asthma and allergy at age 13. Am. J. Respir. Crit. Care Med. 171:137-141. [DOI] [PubMed] [Google Scholar]
  • 128.Simoes, E. A. 1995. Natural and vaccine induced immunity to RSV in primates. NIH publication no. R01 AI37271-02. National Institutes of Health, Bethesda, MD.
  • 129.Simoes, E. A., J. R. Groothuis, X. Carbonell-Estrany, C. H. Rieger, I. Mitchell, L. M. Fredrick, and J. L. Kimpen. 2007. Palivizumab prophylaxis, respiratory syncytial virus, and subsequent recurrent wheezing. J. Pediatr. 151:34-42, 42.e1. [DOI] [PubMed] [Google Scholar]
  • 130.Spann, K. M., K. C. Tran, B. Chi, R. L. Rabin, and P. L. Collins. 2004. Suppression of the induction of alpha, beta, and lambda interferons by the NS1 and NS2 proteins of human respiratory syncytial virus in human epithelial cells and macrophages. J. Virol. 78:4363-4369. (Erratum, 78:6705, 2005.) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Stein, R. T., D. Sherrill, W. J. Morgan, C. J. Holberg, M. Halonen, L. M. Taussig, A. L. Wright, and F. D. Martinez. 1999. Respiratory syncytial virus in early life and risk of wheeze and allergy by age 13 years. Lancet 354:541-545. [DOI] [PubMed] [Google Scholar]
  • 132.Sudo, K., W. Watanabe, S. Mori, K. Konno, S. Shigeta, and T. Yokota. 1999. Mouse model of respiratory syncytial virus infection to evaluate antiviral activity in vivo. Antivir. Chem. Chemother. 10:135-139. [DOI] [PubMed] [Google Scholar]
  • 133.Tekkanat, K. K., H. F. Maassab, D. S. Cho, J. J. Lai, A. John, A. Berlin, M. H. Kaplan, and N. W. Lukacs. 2001. IL-13-induced airway hyperreactivity during respiratory syncytial virus infection is STAT6 dependent. J. Immunol. 166:3542-3548. [DOI] [PubMed] [Google Scholar]
  • 134.Thomas, L. H., J. S. Friedland, M. Sharland, and S. Becker. 1998. Respiratory syncytial virus-induced RANTES production from human bronchial epithelial cells is dependent on nuclear factor-kappa B nuclear binding and is inhibited by adenovirus-mediated expression of inhibitor of kappa B alpha. J. Immunol. 161:1007-1016. [PubMed] [Google Scholar]
  • 135.Tian, B., Y. Zhang, B. A. Luxon, R. P. Garofalo, A. Casola, M. Sinha, and A. R. Brasier. 2002. Identification of NF-κB-dependent gene networks in respiratory syncytial virus-infected cells. J. Virol. 76:6800-6814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Tripp, R. A., D. Moore, A. T. Barskey, L. Jones, C. Moscatiello, H. Keyserling, and L. J. Anderson. 2002. Peripheral blood mononuclear cells from infants hospitalized because of respiratory syncytial virus infection express T helper-1 and T helper-2 cytokines and CC chemokine messenger RNA. J. Infect. Dis. 185:1388-1394. [DOI] [PubMed] [Google Scholar]
  • 137.Vicente, D., M. Montes, G. Cilla, E. G. Perez-Yarza, and E. Perez-Trallero. 2003. Hospitalization for respiratory syncytial virus in the paediatric population in Spain. Epidemiol. Infect. 131:867-872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Wedde-Beer, K., C. Hu, M. M. Rodriguez, and G. Piedimonte. 2002. Leukotrienes mediate neurogenic inflammation in lungs of young rats infected with respiratory syncytial virus. Am. J. Physiol. Lung Cell. Mol. Physiol. 282:L1143-L1150. [DOI] [PubMed] [Google Scholar]
  • 139.Welliver, R. C. 2003. Review of epidemiology and clinical risk factors for severe respiratory syncytial virus (RSV) infection. J. Pediatr. 143:S112-S117. [DOI] [PubMed] [Google Scholar]
  • 140.Welliver, R. C., II, K. H. Hintz, M. Glori, and R. C. Welliver, Sr. 2003. Zileuton reduces respiratory illness and lung inflammation, during respiratory syncytial virus infection, in mice. J. Infect. Dis. 187:1773-1779. [DOI] [PubMed] [Google Scholar]
  • 141.Welliver, R. C., and L. Duffy. 1993. The relationship of RSV-specific immunoglobulin E antibody responses in infancy, recurrent wheezing, and pulmonary function at age 7-8 years. Pediatr. Pulmonol. 15:19-27. [DOI] [PubMed] [Google Scholar]
  • 142.Yu, Q., R. W. Hardy, and G. W. Wertz. 1995. Functional cDNA clones of the human respiratory syncytial (RS) virus N, P, and L proteins support replication of RS virus genomic RNA analogs and define minimal trans-acting requirements for RNA replication. J. Virol. 69:2412-2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zhang, W. D., H. Yang, X. Y. Kong, S. Mohapatra, H. San Juan-Vergara, G. Hellermann, S. Behera, R. Singam, R. F. Lockey, and S. S. Mohapatra. 2005. Inhibition of respiratory syncytial virus infection with intranasal siRNA nanoparticles targeting the viral NS1 gene. Nat. Med. 11:56-62. [DOI] [PubMed] [Google Scholar]
  • 144.Zhang, Y., B. A. Luxon, A. Casola, R. P. Garofalo, M. Jamaluddin, and A. R. Brasier. 2001. Expression of respiratory syncytial virus-induced chemokine gene networks in lower airway epithelial cells revealed by cDNA microarrays. J. Virol. 75:9044-9058. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Microbiology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES