You are on page 1of 10

Infectious Disease–Review

Veterinary Pathology
2015, Vol. 52(1) 97-106
The Relationship Between Respiratory ª The Author(s) 2014
Reprints and permission:
sagepub.com/journalsPermissions.nav
Syncytial Virus and Asthma DOI: 10.1177/0300985814520639
vet.sagepub.com

C. J. Knudson1 and S. M. Varga1,2,3

Abstract
Asthma is a chronic inflammatory disease of the lung that is a leading cause of morbidity and mortality in children worldwide. Most
infants who experience wheezing episodes also exhibit evidence of an ongoing respiratory viral infection. Respiratory syncytial
virus (RSV) is the leading cause of lower respiratory tract infection in children and is a common cause of wheezing in infants and
young children. In the past several decades, a number of studies have demonstrated a relationship between infants with severe
RSV infections and the subsequent development of asthma later during childhood. This review provides an overview of data that
suggests a severe RSV infection early in childhood is linked to development of asthma later in life. In addition, the current and
potential future use of various animal models to gain additional insight into the relationship between RSV and asthma is discussed.

Keywords
asthma, bronchiolitis, respiratory syncytial virus, wheezing, animal models, Th2 cells, Th17 cells, immunoregulation

Asthma is a common chronic inflammatory disease of the respira- once.22 While RSV is a significant health care burden, there
tory tract that is characterized by narrowing of the airways that is currently no licensed vaccine available. Over the past 20
recurs throughout an individual’s lifetime. Prevalence rates for years, there have been a number of studies demonstrating a pos-
asthma are at historically high levels and remain a significant pub- itive correlation between severe RSV infection early in child-
lic health problem.2 Asthma attacks are often characterized by hood and a greater risk of developing asthma.29,53,55,80,81
recurring episodes of symptoms such as coughing, wheezing, This review explores the relationship between severe RSV
shortness of breath, and chest pain. Acute asthma episodes can infection and the subsequent development of asthma. Further-
be triggered by a number of factors, including allergens, chemical more, it describes the use of BALB/c mouse models to explore
irritants, drugs, infection, strenuous exercise, or sudden changes this relationship and discusses the potential utility of other ani-
in weather. It is currently unclear what factors lead to the initial mal models of RSV and asthma that could be used to shed fur-
development of asthma. It is likely that asthma inception is due ther light on this important topic.
to a combination of both genetic predisposition and environmen-
tal cues. Virus infection may represent a critical environmental
Relationship Between Severe RSV Infection
trigger for the development of asthma. RNA from common
respiratory viral infections can be detected in 90% of nasal lavage and Asthma
samples from children aged 1 to 3 years experiencing wheezing Epidemiology
episodes.35 Rhinovirus and respiratory syncytial virus (RSV) are
the most common viruses detected in children experiencing RSV is a seasonal virus in temperate climates with infection
wheezing.35 A study of children with wheezing symptoms rates peaking during winter months. However, in tropical cli-
revealed a strong correlation with a concurrent rhinovirus and/ mates, RSV infection can occur year-round.8 While virtually
or RSV infection. Children with wheezing episodes who were all children eventually become infected with RSV, only 0.5%
infected by either rhinovirus or RSV in the first 3 years of life had to 2% of infants require hospitalization during their initial
a greater tendency to develop asthma by age 6 years compared
with children not infected by either virus.35 This suggests that 1
Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa
exposure to common respiratory viral infections such as RSV dur- City, IA, USA
2
ing early childhood may increase the risk of developing asthma. Department of Microbiology, University of Iowa, Iowa City, IA, USA
3
RSV is a negative-sense single-stranded RNA virus of the Department of Pathology, University of Iowa, Iowa City, IA, USA
Paramyxoviridae family and a common cause of bronchiolitis
Corresponding Author:
in infants. RSV is the leading cause of lower respiratory tract S. M. Varga, University of Iowa, 51 Newton Rd, 3-532 BSB, Iowa City, IA 52242,
infections in infants and young children.27,77,93 By the age 2 USA.
years, nearly all children have been infected by RSV at least Email: steven-varga@uiowa.edu
98 Veterinary Pathology 52(1)

exposure.76 Hospitalizations during RSV infection are a result time of year at which there was the greatest number of
of airway dysfunction caused by inflammation of the bronch- bronchiolitis-related hospitalizations (BRHs). The number of
ioles resulting in bronchiolitis. Reinfection by RSV can occur BRHs peaks every year during the winter months in temperate
throughout one’s lifetime as sterilizing immunity is not gener- climates. If there was no relationship between the risk of devel-
ated following infection.22,25,28 However, subsequent RSV oping asthma and severe bronchiolitis, then rates of asthma
exposures are believed to be less severe in healthy adults. among children would be expected to be similar year-round
irrespective of patterns of BRH. However, the authors observed
similar patterns between the timing of the peak of BRHs and
Link Between Severe RSV Infection and Development of the likelihood of infants developing asthma. Children born 4
Recurrent Wheezing or Asthma months prior to the annual peak of BRHs were 29% more likely
A large number of studies have identified a link between severe to develop asthma compared with infants born 1 year from this
RSV infection and an increased likelihood to develop either time point. This relationship was similar across the 5-year
recurrent wheezing or asthma.* The Avon Longitudinal Study study even though the peak of BRHs shifted by up to 6 weeks.
of Parents and Children by Henderson et al29 revealed that chil- It was not determined if children were infected with a specific
dren admitted to the hospital for RSV-induced bronchiolitis virus such as RSV, although up to 70% of severe bronchiolitis
prior to the age 1 year were at increased risk of developing cases are associated with RSV infection.58 The TABS suggests
recurrent wheezing at 30 to 42 and 69 to 81 months compared that viral infection during childhood directly contributes to an
with the control group (28.1% vs 13.1% and 22.6% vs 9.6%, increased risk of developing asthma.
respectively). Furthermore, 38.4% of the severe RSV group
developed asthma by age 7 years compared with only 20.1% Influence of Age in Longitudinal RSV and Asthma Studies
of the control group. Similar results were observed in a long-
term Swedish study of 140 children by Sigurs et al.80 Children Although a number of studies indicate an increased risk of
who experienced a severe RSV infection prior to 1 year of age developing recurrent wheezing later during childhood follow-
were at a much greater risk of developing asthma by age 7.5 ing a severe RSV infection, a few longitudinal studies suggest
years (30% vs 3% of control group). In addition, recurrent these changes are transient. A study by Pullan and Hey65 deter-
wheezing symptoms were more likely to occur in the children mined that children with severe RSV infections prior to 1 year
who experienced RSV-induced bronchiolitis (28%) compared of age experienced recurrent wheezing symptoms primarily
with the control group (11%). A 5-year and a 10-year follow- during the first 4 years of life (38% vs 15% of control group).
up study of the same cohort revealed similar trends. Children However, by age 10 years, there was no longer a significant dif-
who had experienced a severe RSV infection during infancy ference in rates of wheezing (6.2% vs 4.5%). Similar results
exhibited a significantly greater incidence of either recurrent were observed in the Tucson Children’s Respiratory Study of
wheezing (30% vs 16.3%) or asthma (37% vs 5.4%) by age 1246 children.87,91 Infants who experienced a severe RSV
13 years.81 The incidence of recurrent wheezing (30% vs infection in the first 3 years of life were at a significantly
1%) remained significantly higher in children with an early increased risk of exhibiting recurrent wheezing symptoms at
severe RSV infection by age 18 years.79 The authors also noted ages 6 and 11 years. However, there was no significant differ-
that children who had experienced an early severe RSV infec- ence in infrequent or frequent wheezing symptoms by age 13
tion were more likely to develop allergic sensitization to com- years. These data are in contrast to the longitudinal study by
mon inhaled or food allergens at any of the 3 age time points Sigurs et al81 that demonstrated a significant increase in chil-
investigated.79–81 Since allergen sensitization is a major risk dren with asthma by age 13 years. The differential findings
factor associated with asthma inception, severe RSV infection reported in these studies could be due to many factors, includ-
during early childhood may contribute to the development of ing genetic variability among cohorts, parameters used to select
asthma through sensitization to common allergens.60,68,84,86 the cohort, how wheezing/asthma is evaluated, and differences
Overall, these studies suggest that experiencing a severe RSV in the relative pathogenicity of circulating RSV strains.
infection during early childhood increases the likelihood of
developing asthma later in life. Palivizumab Prophylaxis Treatment and the
While numerous studies indicate a link between early severe Development of Recurrent Wheezing
RSV infection and an increased risk in developing asthma, the
exact nature of the relationship is unclear. The results from the An alternative approach to determining if RSV contributes to
Tennessee Asthma Bronchiolitis Study (TABS) of 95 310 chil- the development of asthma is to limit RSV-induced disease.
dren by Carroll et al10 and Wu et al98 suggest that a causal rela- Although there is currently no licensed vaccine, passive immu-
tionship exists between severe RSV infection and the nization has shown some promise.24,69 In the mid-1990s, a
development of asthma. This study assessed the relationship monoclonal antibody specific for the fusion protein of RSV,
between the risk of developing asthma by age 5.5 years and the palivizumab, was evaluated across the United States, United
Kingdom, and Canada for efficacy in the prophylactic treat-
ment of RSV infection.1 Palivizumab prophylaxis reduced
*References 18, 29, 37, 39, 47, 53, 55, 80, 81, 87, 98. RSV-associated hospitalization rates by 55% (10.6% placebo
Knudson and Varga 99

vs 4.8% palivizumab). Due to cost considerations, palivizu- with RSV by age 2 years revealed significantly elevated IL-6
mab prophylaxis is administered only to children at high risk protein levels compared with controls.56 Furthermore, in mur-
of developing severe RSV-induced disease, such as preterm ine models of allergic inflammatory disease, protein levels of
infants. In a study by Simoes et al,82 preterm infants who proinflammatory chemokines MIP-1a and CCL5 (RANTES)
either had received or had never been administered palivizu- were elevated and significantly contributed to allergic airway
mab prophylaxis were followed for development of wheezing inflammation.36,71 Increased levels of RANTES protein also
symptoms. have been correlated with increased RSV disease severity.
This study indicated there were long-term benefits to lessen- Studies also suggest that an imbalance in the type of CD4
ing the severity of RSV infection. The rates of recurrent wheez- T-cell response further exacerbates allergic responses. Infec-
ing were compared between infants with prophylactic treatment tion by RSV at an early age tends to induce a type 2 helper
who were not hospitalized for severe RSV infection and children T-cell response (Th2) that produces IL-4, IL-5, and IL-13.6,14
who were hospitalized for RSV-induced bronchiolitis without A Th2-biased immune response is known to contribute to dis-
prior palivizumab treatment. Children from the prophylaxis ease in murine models of allergic airway inflammation and is
group were significantly less likely to develop physician- strongly associated with asthmatic patients.13,17,32,73,78 There-
diagnosed recurrent wheezing (8% vs 16% of no prophylaxis fore, infection by RSV at an early age may induce a Th2-
group) by 3.5 years of age.82 These data further support the biased immune response that promotes asthma inception.
notion that severe RSV infection may contribute to the develop- Recent work also suggests a role for IL-17–induced patho-
ment of asthma. genesis in a subset of asthmatic patients. IL-17 is secreted by
type 17 helper T cells (Th17) and can induce the chemotaxis
of granulocytes such as neutrophils or directly signal through
Lung Function and Immune Response to cells such as fibroblasts and macrophages to produce proin-
RSV Infection flammatory cytokines IL-1b, IL-6, and TNF-a.51 Levels of
IL-17 are elevated in the sputum and bronchoalveolar lavage
Lung Function (BAL) of asthmatic human patients.15,51 A Th17-biased immune
Infants younger than 6 months are highly susceptible to RSV- response and subsequent neutrophilia is also observed in mice
induced bronchiolitis.21,59 This is likely due to their reduced following allergic sensitization to ovalbumin (OVA).97 Further-
pulmonary function since lung development continues for sev- more, both IL-17 secretion and neutrophil influx were necessary
eral years.49,100 Length of hospital stay often correlates with the to mediate granulocytic chemotaxis, mucus hypersecretion, and
degree of lung dysfunction, and a significant number of infants airway hypersensitivity following allergic sensitization.51,97
with severe RSV-induced disease require mechanical ventila- These studies indicate that Th17 responses play a critical role
tion.90 If severe RSV infection increases an individual’s risk in mediating particular forms of asthma. In addition, levels of
of asthma inception, there may be long-term alterations to the IL-17 are elevated in both humans and mice with severe RSV
lung environment. In agreement, children evaluated approxi- infection.52 Neutralization of IL-17 signaling during RSV infec-
mately 6 years after a severe RSV infection exhibited signifi- tion significantly reduced granulocytic infiltrate, airway resis-
cantly reduced respiratory capacity compared with infants tance, and mucus hypersecretion.52 These data suggest that
who were not hospitalized for RSV infection.63 A study by elevated Th17 responses following RSV infection may increase
Korppi et al37 also demonstrated reduced lung function in an individual’s susceptibility to developing asthma.
young adults 20 years after a severe RSV infection. Similar
results were observed in a number of other studies in which
children exhibited reduced lung function 7 or more years after
a severe RSV infection.11,41,50,55,81,85 These studies indicate Genetic Predisposition to Asthma and the
that severe RSV infection may alter the lung environment and Influence of RSV Infection
lead to prolonged lung dysfunction long after the infection is Two-Hit Hypothesis
resolved.
It is possible that children who are genetically predisposed to
developing asthma will eventually develop chronic lung
Immune Response inflammation and are more likely to experience greater mor-
Infection with RSV results in a significant increase in proin- bidity from RSV infection. Children who exhibit bronchiolitis
flammatory cytokines and chemokines such as interleukin prior to 1 year of age show signs of reduced lung function at 5
(IL)–1a, IL-1b, IL-6, IL-8, interferon (IFN)–g, tumor necrosis weeks old.100 However, as discussed, a number of studies sug-
factor (TNF)–a, monocyte chemotactic protein 1 (MCP-1), gest that severe RSV infection may increase an individual’s
macrophage inflammatory protein (MIP)–1a, and MIP-1b. risk of developing asthma later in life. Children younger than
Since asthma is a chronic inflammatory disease, severe RSV 6 months exhibit the greatest risk of severe morbidity from
infection may promote a prolonged inflammatory environment an RSV infection.21,59 It is unclear how RSV infection would
within the lung that leads to the development of asthma. Naso- promote asthma, a chronic lung disease, years after the primary
pharyngeal aspirates obtained from asthmatic infants infected infection has been resolved.
100 Veterinary Pathology 52(1)

It has been proposed that the development of asthma may severe morbidity during RSV infection and an increased likeli-
require a ‘‘2-hit’’ scenario as illustrated in Fig. 1.31,42 This hood to develop recurrent wheezing.94 Furthermore, children
hypothesis suggests that asthma inception depends on the inter- with elevated CCL5 levels from nasal epithelium at the time
action of at least 2 genetic, developmental, or environmental fac- of RSV-induced bronchiolitis exhibit an increased risk of
tors. An example may be a polymorphism in a cytokine that developing asthma by age 7 years.5
predisposes an individual to develop allergic responses. If an Attachment of the RSV virion to the host epithelial cell is
individual with this polymorphism experiences a severe RSV mediated through the G protein of RSV, which is necessary for
infection, he or she is more likely to exhibit either long-term optimal infection.43,92 While the G protein of RSV typically
symptoms or chronic inflammation such as asthma. The 2-hit binds to highly sulfated heparin-like glucosaminoglycans, it
hypothesis also predicts that if only one factor is present, then also contains a chemokine motif similar to CX3CL1 or frac-
either disease or allergic responses would be transient. This may talkine.19,96 The CX3CL1 mimicry function of RSV likely
explain why not all children who experience severe RSV- allows the virus to alter the host immune response.12,96 A
induced bronchiolitis develop asthma as well as why some chil- T280M mutation in CX3CR1, which encodes for the receptor
dren with early childhood wheezing exhibit resolution of their that binds CX3CL1, has also been associated with increased
illness by their adolescence.46,65,87,88 A number of gene poly- morbidity following RSV infection and an increased risk of
morphisms are associated with both asthma and susceptibility developing asthma.4,95 These studies suggest that excessive
to severe RSV-induced disease, as discussed below. CX3CL1-CX3CR1 signaling following RSV infection in chil-
dren with CX3CR1 polymorphisms further predisposes those
infants to asthma inception.
Cytokines IL-8 is a chemokine secreted by epithelial cells and macro-
Cytokines play a significant role in influencing the type of phages that recruits primarily neutrophils but also targets
immune response that develops following infection. Cytokines macrophages, mast cells, endothelial cells, and keratinocytes.
such as IL-4 and IL-13 are primarily produced by Th2 CD4 T Therefore, IL-8 is an important mediator of the innate immune
cells and play a crucial role in the induction of some allergic system to initiate inflammation. Two separate studies by Hull
immune responses. Children with gain-of-function polymorph- et al33,34 demonstrate that gain-of-function polymorphisms in
isms in both IL4RA and IL13 are almost 5 times as likely to the promoter region of IL8 lead to increased disease severity
develop asthma compared to individuals without polymorphisms in infants following RSV infection. Polymorphisms in IL-8 are
in either gene.32 Gain-of-function polymorphisms in IL-4 and also associated with an increased risk of developing asthma.67
IL-13 cytokine genes have also been reported to be increased Furthermore, a study of 164 children in the United Kingdom
in children hospitalized for RSV infection.30,66 A study by revealed that children with a IL-8 polymorphism were signifi-
Ermers et al17 also determined that following a severe RSV cantly more likely to have episodes of wheezing following
infection, children with the IL13-R130Q polymorphism were RSV-related bronchiolitis.23 Taken together, these studies sug-
significantly more likely to experience wheezing at age 6 years. gest that genetic predispositions to allergic illness can alter the
In addition, children with low IFN-g, a type 1 helper CD4 T-cell disease severity of RSV infection and the subsequent develop-
(Th1)–associated cytokine, also exhibited an increased risk in ment of asthma.
developing recurrent wheezing during infancy.20,89 Taken
together, these studies indicate that an imbalanced immune sys-
tem, Th2 biased over Th1, may contribute to severe RSV infec-
Immune Tolerance
tion and subsequent asthma inception. RSV infection may also increase the susceptibility to develop
allergic immune responses by breaking immune tolerance to
allergens early in life. Regulatory T cells (Tregs) play an essen-
Chemokines tial role in mediating immune tolerance and prevention of
Chemokines are critical for the recruitment and adhesion of immune responses to allergens.44 Tregs can regulate the immu-
leukocytes during infection. Polymorphisms in either chemo- nogenicity of dendritic cells (DCs) by reducing DC expression
kines or their receptors may predispose an individual toward of the costimulatory molecules CD80 and CD86 as well as
chronic pulmonary inflammation and increase disease severity expression of major histocompatibility complex (MHC) class
following respiratory viral infection. The chemokine CCL5, II.44 However, infection of mice with RSV following OVA
also known as RANTES, is an important chemotactic agent for sensitization significantly inhibits the suppressive capacity of
Th2-associated immune cells, including Th2 cells, eosinophils, Tregs.38 The impaired Treg response correlates with increased
and basophils. Gain-of-function polymorphisms in the promo- DC expression of the costimulatory molecules CD40 and CD86
ter region of the CCL5 gene have been associated with asthma as well as MHC class II. This suggests that if immune tolerance
and severe airway dysfunction.40 These promoter region poly- is broken by RSV infection, infants may be more susceptible to
morphisms have also been correlated with severe cases of the development of chronic inflammation to allergens and sub-
RSV-induced bronchiolitis.3 A study of 320 Chinese children sequent asthma. Furthermore, RSV infection may differentially
with RSV-induced bronchiolitis revealed an increased risk of affect the function of various DC subsets. RSV infection of
infants with CCL5 promoter region polymorphisms to exhibit myeloid DCs, but not plasmacytoid DCs, leads to the
Knudson and Varga 101

Figure 1. Proposed model of severe respiratory syncytial virus (RSV) infection and asthma inception based on the ‘‘2-hit’’ hypothesis. A young
infant must have a combination of 1 or more genetic factor(s) and RSV-related immunological factor(s) to develop asthma as suggested by the
2-hit hypothesis. An infant with a genetic predisposition to allergen sensitization can develop asthma following a severe RSV infection. For exam-
ple, an infant with a gain-of-function gene polymorphism for the cytokine interleukin (IL)–4 may exhibit a Th2-biased immune response following
a severe RSV infection. In the absence of either a genetic factor or a severe RSV infection, thus not fulfilling a 2-hit scenario, the infant will likely
experience either transient pulmonary dysfunction or no disease at all. A child with a gain-of-function CCL5 polymorphism who experiences a
mild RSV infection is an example of this, likely having either short-term pulmonary dysfunction or no long-term disease. Last, an infant who
experiences a mild or asymptomatic RSV infection with no genetic predisposition toward allergic sensitization will exhibit no long-term disease.

upregulation of maturation markers and an increased capacity are important for promoting allergic sensitization, whereas
to induce the proliferation of OVA-specific T cells in vitro.7 plasmacytoid DCs are tolerogenic.57 Together, these studies
Previous work has indicated that immunogenic myeloid DCs suggest that impaired regulation of allergic immune responses
102 Veterinary Pathology 52(1)

asthma.62,75 BALB/c mice can be infected with RSV either


prior to or following allergic sensitization to OVA, as illu-
strated in Fig. 2A and Fig. 2B, respectively. Both models lead
to increased airway resistance in comparison to OVA-
sensitized mice not infected with RSV. Studies using these 2
models have yielded several significant pieces of information.
Using the model of allergic sensitization followed by RSV
infection, Peebles et al62 reported that RSV-infected mice exhi-
bit greater airway resistance for up to 2 weeks following infec-
tion compared with the OVA sensitization alone or acute RSV
infection groups. Interestingly, the elevated airway hyperreac-
tivity did not correlate with an elevated Th2 immune response
since Th2 CD4 T cells and Th2 cytokine protein levels were
similar to the OVA sensitization alone group.61 In contrast, the
Th2-associated cytokines IL-4 and IL-5 play a crucial role in
mediating the elevated airway hyperresponsiveness in the mur-
ine model of RSV infection followed by OVA sensitization.74
Similar results were observed by Lukacs et al45 in BALB/c
mice infected with RSV prior to cockroach antigen (CRA) sen-
sitization, as illustrated in Fig. 2C. Neutralization of the Th2-
associated cytokine IL-13 in BALB/c mice sensitized against
CRA following RSV infection significantly reduced airway
hyperreactivity. Furthermore, mice lacking IL-17A signaling
exhibited significantly greater numbers of eosinophils and lym-
phocytes as well as increased levels of IL-13 protein in the lung
following RSV infection of OVA-sensitized mice.54 In addi-
tion, the IL-17A–deficient mice had significantly elevated air-
way resistance compared with wild-type (WT) mice. These
data suggest that, while Th2-biased immune responses may
be critical to mediate RSV-induced inception of asthma,
Th17 responses limit airway hyperreactivity potentially by
Figure 2. Murine models of allergen sensitization and respiratory syn- inhibiting a pathogenic Th2 immune response.
cytial virus (RSV) infection. (A) BALB/c mice are sensitized to OVA The development and use of alternative animal models is a
following RSV challenge and analyzed on day 21 postinfection (PI).75
(B) BALB/c mice are sensitized to OVA prior to RSV challenge and crucial step to further investigate the relationship between
assessed on day 15 PI.62 (C) BALB/c mice are infected with RSV prior severe RSV infection and asthma. While mice are an appealing
to sensitization against cockroach antigen (CRA) and challenged with model due to their availability and relatively low cost, mice are
methacholine 43 days PI.45 (D) Cotton rats are sensitized to Aspergillus semi-permissive for RSV replication. In contrast, cotton rats
fumigatus (Af) and infected with RSV 14 days later. Cotton rats are exhibit approximately 100-fold greater peak viral titers com-
challenged with Af on day 2 PI and airway hyperreactivity assessed pared with BALB/c mice following RSV infection but do not
on day 4 PI.26 i.n., intranasal; i.p., intraperitoneal; i.t., intratracheal. show overt clinical signs of illness.64,70 A study by Hassantou-
fighi et al26 has established a cotton rat model to investigate the
following RSV infection can contribute to the development of relationship between RSV and asthma as outlined in Fig. 2D.
asthma. Infection of cotton rats with RSV following sensitization to the
fungus Aspergillus fumigatus (Af) induces upregulation of IL-4,
IL-5, and IL-13 as well as a significant increase in airway
Use of Murine Models to Explore the
hyperreactivity following methacholine challenge. Further-
Relationship Between RSV and Asthma more, treatment of the Af-sensitized cotton rats with palivizu-
While many human studies have indicated a role for RSV- mab 2 days following RSV infection significantly reduced
induced inception of asthma, far less work has been performed airway hyperreactivity. This initial study indicates that RSV
using animal models to explore the underlying immunological exacerbates allergic immune responses and that prophylactic
mechanisms that link RSV infection and the induction of treatment to prevent RSV infection may help either prevent
asthma. This is due to the complexity of asthma and the lack or reduce the severity of asthma.
of animal models that reflect the entire disease spectrum Nonhuman primates, specifically macaque monkeys, repre-
observed in RSV-infected humans.9,16 However, murine mod- sent another animal model for RSV infection and show mild signs
els of allergen sensitization have been used extensively to of clinical illness.48,83 In addition, rhesus monkeys (Macaca
investigate the impact of RSV infection in the induction of mulatta) experience elevated airway hyperresponsiveness and
Knudson and Varga 103

increased plasma IgE following sensitization to house dust mite Declaration of Conflicting Interests
antigen and subsequent allergen rechallenge.72,99 However, there The author(s) declared no potential conflicts of interest with respect to
has been no investigation to determine the impact of RSV infec- the research, authorship, and/or publication of this article.
tion on allergic immune responses in nonhuman primates. Due to
the high degree of genetic similarity between humans and mon- Funding
keys, establishment of a nonhuman primate model to investigate The author(s) received no financial support for the research, author-
the role of RSV infection on the development of asthma would be ship, and/or publication of this article.
of great benefit.
References
1. Palivizumab, a humanized respiratory syncytial virus monoclonal
antibody, reduces hospitalization from respiratory syncytial virus
Summary infection in high-risk infants. Pediatrics. 1998;102(3):531–537.
RSV infection is the leading cause of lower respiratory tract 2. Akinbami L. The state of childhood asthma, United States, 1980–
infections in children worldwide. It is a significant cause of 2005. Adv Data. 2006;(381):1–24.
morbidity and mortality in infants. To date, there are no 3. Amanatidou V, Sourvinos G, Apostolakis S, et al. RANTES pro-
licensed vaccines for RSV, and only limited supportive care moter gene polymorphisms and susceptibility to severe respira-
is provided for severe cases. A number of studies have tory syncytial virus–induced bronchiolitis. Pediatr Infect Dis J.
established a strong correlation between severe RSV infec- 2008;27(1):38–42.
tion and the development of asthma during childhood. 4. Amanatidou V, Sourvinos G, Apostolakis S, et al. T280 M varia-
While genetic mutations may contribute to susceptibility tion of the CX3C receptor gene is associated with increased risk
toward asthma inception, it is highly likely that other envi- for severe respiratory syncytial virus bronchiolitis. Pediatr Infect
ronmental factors such as an early RSV infection may act as Dis J. 2006;25(5):410–414.
an initial trigger event. However, additional studies are 5. Bacharier LB, Cohen R, Schweiger T, et al. Determinants of
required to elucidate the underlying immunological mechan- asthma after severe respiratory syncytial virus bronchiolitis.
isms of how RSV infection contributes to the subsequent J Allergy Clin Immunol. 2012;130(1):91–100.e103.
induction of asthma. These studies must take into consider- 6. Barends M, de Rond LG, Dormans J, et al. Respiratory syncytial
ation the use of large cohorts over several decades with pre- virus, pneumonia virus of mice, and influenza A virus differently
cise assessment of any genetic predispositions to accurately affect respiratory allergy in mice. Clin Exp Allergy. 2004;34(3):
determine if early severe RSV infection has a causal rela- 488–496.
tionship to the development of asthma. Further use of pro- 7. Boogaard I, van Oosten M, van Rijt LS, et al. Respiratory syncy-
phylactic treatment with palivizumab may also help tial virus differentially activates murine myeloid and plasmacy-
determine if infants with a genetic predisposition to allergic toid dendritic cells. Immunology. 2007;122(1):65–72.
responses still develop asthma independent of having a 8. Brandenburg AH, Neijens HJ, Osterhaus AD. Pathogenesis of
severe RSV infection early in childhood. Overall, these RSV lower respiratory tract infection: implications for vaccine
studies would help ascertain whether RSV prophylaxis is a development. Vaccine. 2001;19(20–22):2769–2782.
viable option to indirectly prevent asthma inception in chil- 9. Byrd LG, Prince GA. Animal models of respiratory syncytial
dren with family histories of such disease. virus infection. Clin Infect Dis. 1997;25(6):1363–1368.
Further investigation of these questions needs to be vigor- 10. Carroll KN, Wu P, Gebretsadik T, et al. Season of infant bronch-
ously pursued using animal models. The accessibility, rela- iolitis and estimates of subsequent risk and burden of early child-
tively low cost of maintenance, and broad availability of hood asthma. J Allergy Clin Immunol. 2009;123(4):964–966.
reagents and genetic mutations in mice will allow for thorough 11. Cassimos DC, Tsalkidis A, Tripsianis GA, et al. Asthma, lung
investigation of all of these questions. While models of allergen function and sensitization in school children with a history of
sensitization and RSV infection have been defined, there has bronchiolitis. Pediatr Int. 2008;50(1):51–56.
been little investigation of the cellular composition and 12. Choi Y, Mason CS, Jones LP, et al. Antibodies to the central con-
changes in pulmonary architecture in these murine models. The served region of respiratory syncytial virus (RSV) G protein block
development of new models in other animals such as cotton rats RSV G protein CX3C-CX3CR1 binding and cross-neutralize
and nonhuman primates would also be beneficial since RSV RSV A and B strains. Viral Immunol. 2012;25(3):193–203.
replicates to a greater extent in these animals compared with 13. Dakhama A, Lee YM, Ohnishi H, et al. Virus-specific IgE
mice. Together, nonhuman models will help tease apart the enhances airway responsiveness on reinfection with respiratory
immunological mechanisms associated with severe RSV infec- syncytial virus in newborn mice. J Allergy Clin Immunol. 2009;
tion that may mediate asthma inception, which cannot be eval- 123(1):138–145.e135.
uated in human studies. Furthermore, not only will this help to 14. Dakhama A, Park JW, Taube C, et al. The enhancement or pre-
determine if RSV prophylaxis/vaccination is beneficial in the vention of airway hyperresponsiveness during reinfection with
prevention of asthma, but it will also allow for the development respiratory syncytial virus is critically dependent on the age at
of novel strategies to limit chronic disease following the devel- first infection and IL-13 production. J Immunol. 2005;175(3):
opment of asthma. 1876–1883.
104 Veterinary Pathology 52(1)

15. Doe C, Bafadhel M, Siddiqui S, et al. Expression of the T helper 30. Hoebee B, Rietveld E, Bont L, et al. Association of severe respira-
17–associated cytokines IL-17A and IL-17F in asthma and tory syncytial virus bronchiolitis with interleukin-4 and interleukin-
COPD. Chest. 2010;138(5):1140–1147. 4 receptor alpha polymorphisms. J Infect Dis. 2003;187(1):2–11.
16. Domachowske JB, Bonville CA, Rosenberg HF. Animal models 31. Holt PG, Upham JW, Sly PD. Contemporaneous maturation of
for studying respiratory syncytial virus infection and its long term immunologic and respiratory functions during early childhood:
effects on lung function. Pediatr Infect Dis J. 2004;23(11)(suppl): implications for development of asthma prevention strategies.
S228–S234. J Allergy Clin Immunol. 2005;116(1):16–25.
17. Ermers MJ, Hoebee B, Hodemaekers HM, et al. IL-13 genetic 32. Howard TD, Koppelman GH, Xu J, et al. Gene-gene interaction in
polymorphism identifies children with late wheezing after asthma: IL4RA and IL13 in a Dutch population with asthma. Am J
respiratory syncytial virus infection. J Allergy Clin Immunol. Hum Genet. 2002;70(1):230–236.
2007;119(5):1086–1091. 33. Hull J, Ackerman H, Isles K, et al. Unusual haplotypic structure of
18. Escobar GJ, Ragins A, Li SX, et al. Recurrent wheezing in the IL8, a susceptibility locus for a common respiratory virus. Am J
third year of life among children born at 32 weeks’ gestation or Hum Genet. 2001;69(2):413–419.
later: relationship to laboratory-confirmed, medically attended 34. Hull J, Thomson A, Kwiatkowski D. Association of respiratory
infection with respiratory syncytial virus during the first year of syncytial virus bronchiolitis with the interleukin 8 gene region
life. Arch Pediatr Adolesc Med. 2010;164(10):915–922. in UK families. Thorax. 2000;55(12):1023–1027.
19. Feldman SA, Hendry RM, Beeler JA. Identification of a linear 35. Jackson DJ, Gangnon RE, Evans MD, et al. Wheezing rhinovirus
heparin binding domain for human respiratory syncytial virus illnesses in early life predict asthma development in high-risk
attachment glycoprotein G. J Virol. 1999;73(8):6610–6617. children. Am J Respir Crit Care Med. 2008;178(7):667–672.
20. Gern JE, Brooks GD, Meyer P, et al. Bidirectional interactions 36. John AE, Berlin AA, Lukacs NW. Respiratory syncytial virus–
between viral respiratory illnesses and cytokine responses in the induced CCL5/RANTES contributes to exacerbation of allergic
first year of life. J Allergy Clin Immunol. 2006;117(1):72–78. airway inflammation. Eur J Immunol. 2003;33(6):1677–1685.
21. Glezen WP, Paredes A, Allison JE, et al. Risk of respiratory syn- 37. Korppi M, Piippo-Savolainen E, Korhonen K, et al. Respiratory
cytial virus infection for infants from low-income families in rela- morbidity: 20 years after RSV infection in infancy. Pediatr Pul-
tionship to age, sex, ethnic group, and maternal antibody level. monol. 2004;38(2):155–160.
J Pediatr. 1981;98(5):708–715. 38. Krishnamoorthy N, Khare A, Oriss TB, et al. Early infection with
22. Glezen WP, Taber LH, Frank AL, et al. Risk of primary infection respiratory syncytial virus impairs regulatory T cell function and
and reinfection with respiratory syncytial virus. Am J Dis Child. increases susceptibility to allergic asthma. Nat Med. 2012;18(10):
1986;140(6):543–546. 1525–1530.
23. Goetghebuer T, Isles K, Moore C, et al. Genetic predisposition to 39. Kusel MM, de Klerk NH, Kebadze T, et al. Early-life respiratory
wheeze following respiratory syncytial virus bronchiolitis. Clin viral infections, atopic sensitization, and risk of subsequent devel-
Exp Allergy. 2004;34(5):801–803. opment of persistent asthma. J Allergy Clin Immunol. 2007;
24. Groothuis JR, Simoes EA, Levin MJ, et al. Prophylactic adminis- 119(5):1105–1110.
tration of respiratory syncytial virus immune globulin to high-risk 40. Lachheb J, Chelbi H, Hamzaoui K, et al. Association between
infants and young children. The Respiratory Syncytial Virus RANTES polymorphisms and asthma severity among Tunisian
Immune Globulin Study Group. N Engl J Med. 1993;329(21): children. Hum Immunol. 2007;68(8):675–680.
1524–1530. 41. Larouch V, Rivard G, Deschesnes F, et al. Asthma and airway
25. Hall CB, Walsh EE, Long CE, et al. Immunity to and frequency of hyper-responsiveness in adults who required hospital admission
reinfection with respiratory syncytial virus. J Infect Dis. 1991; for bronchiolitis in early childhood. Respir Med. 2000;94(3):
163(4):693–698. 288–294.
26. Hassantoufighi A, Oglesbee M, Richter BW, et al. Respiratory 42. Lemanske RF Jr. The Childhood Origins of Asthma (COAST)
syncytial virus replication is prolonged by a concomitant allergic study. Pediatr Allergy Immunol. 2002;13(suppl 15):38–43.
response. Clin Exp Immunol. 2007;148(2):218–229. 43. Levine S, Klaiber-Franco R, Paradiso PR. Demonstration that gly-
27. Heilman CA. From the National Institute of Allergy and Infec- coprotein G is the attachment protein of respiratory syncytial
tious Diseases and the World Health Organization. Respiratory virus. J Gen Virol. 1987;68(pt 9):2521–2524.
syncytial and parainfluenza viruses. J Infect Dis. 1990;161(3): 44. Lewkowich IP, Herman NS, Schleifer KW, et al. CD4þCD25þ T
402–406. cells protect against experimentally induced asthma and alter pul-
28. Henderson FW, Collier AM, Clyde WA Jr, et al. Respiratory- monary dendritic cell phenotype and function. J Exp Med. 2005;
syncytial-virus infections, reinfections and immunity: a prospec- 202(11):1549–1561.
tive, longitudinal study in young children. N Engl J Med. 1979; 45. Lukacs NW, Tekkanat KK, Berlin A, et al. Respiratory syncy-
300(10):530–534. tial virus predisposes mice to augmented allergic airway
29. Henderson J, Hilliard TN, Sherriff A, et al. Hospitalization for responses via IL-13–mediated mechanisms. J Immunol. 2001;
RSV bronchiolitis before 12 months of age and subsequent 167(2):1060–1065.
asthma, atopy and wheeze: a longitudinal birth cohort study. 46. Mallia P, Johnston SL. Respiratory viruses: do they protect from
Pediatr Allergy Immunol. 2005;16(5):386–392. or induce asthma? Allergy. 2002;57(12):1118–1129.
Knudson and Varga 105

47. Martinez FD, Wright AL, Taussig LM, et al. Asthma and wheez- infection in early childhood in Guinea-bissau. Pediatr Infect
ing in the first six years of life. The Group Health Medical Associ- Dis J. 2006;25(11):1025–1031.
ates. N Engl J Med. 1995;332(3):133–138. 64. Prince GA, Jenson AB, Horswood RL, et al. The pathogenesis of
48. McArthur-Vaughan K, Gershwin LJ. A rhesus monkey model of respiratory syncytial virus infection in cotton rats. Am J Pathol.
respiratory syncytial virus infection. J Med Primatol. 2002;31(2): 1978;93(3):771–791.
61–73. 65. Pullan CR, Hey EN. Wheezing, asthma, and pulmonary dysfunc-
49. Merkus PJ, ten Have-Opbroek AA, Quanjer PH. Human lung tion 10 years after infection with respiratory syncytial virus in
growth: a review. Pediatr Pulmonol. 1996;21(6):383–397. infancy. Br Med J (Clin Res Ed). 1982;284(6330):1665–1669.
50. Mok JY, Simpson H. Outcome of acute lower respiratory tract 66. Puthothu B, Krueger M, Forster J, et al. Association between
infection in infants: preliminary report of seven-year follow-up severe respiratory syncytial virus infection and IL13/IL4 haplo-
study. Br Med J (Clin Res Ed). 1982;285(6338):333–337. types. J Infect Dis. 2006;193(3):438–441.
51. Molet S, Hamid Q, Davoine F, et al. IL-17 is increased in asth- 67. Puthothu B, Krueger M, Heinze J, et al. Impact of IL8 and
matic airways and induces human bronchial fibroblasts to produce IL8-receptor alpha polymorphisms on the genetics of bron-
cytokines. J Allergy Clin Immunol. 2001;108(3):430–438. chial asthma and severe RSV infections. Clin Mol Allergy.
52. Mukherjee S, Lindell DM, Berlin AA, et al. IL-17–induced pul- 2006;4:2.
monary pathogenesis during respiratory viral infection and 68. Remes ST, Koskela HO, Iivanainen K, et al. Allergen-specific
exacerbation of allergic disease. Am J Pathol. 2011;179(1): sensitization in asthma and allergic diseases in children: the study
248–258. on farmers’ and non-farmers’ children. Clin Exp Allergy. 2005;
53. Murray M, Webb MS, O’Callaghan C, et al. Respiratory status 35(2):160–166.
and allergy after bronchiolitis. Arch Dis Child. 1992;67(4): 69. Rodriguez WJ, Gruber WC, Groothuis JR, et al. Respiratory syn-
482–487. cytial virus immune globulin treatment of RSV lower respiratory
54. Newcomb DC, Boswell MG, Reiss S, et al. IL-17A inhibits airway tract infection in previously healthy children. Pediatrics. 1997;
reactivity induced by respiratory syncytial virus infection during 100(6):937–942.
allergic airway inflammation. Thorax. 2013;68(8):717–723. 70. Sadowski W, Wilczynski J, Semkow R, et al. The cotton rat
55. Noble V, Murray M, Webb MS, et al. Respiratory status and (Sigmodon hispidus) as an experimental model for studying viruses
allergy nine to 10 years after acute bronchiolitis. Arch Dis Child. in respiratory tract infections, II: influenza viruses types A and B
1997;76(4):315–319. [in Polish]. Med Dosw Mikrobiol. 1987;39(1):43–55.
56. Oh JW, Lee HB, Park IK, et al. Interleukin-6, interleukin-8, 71. Schaller MA, Kallal LE, Lukacs NW. A key role for CC chemo-
interleukin-11, and interferon-gamma levels in nasopharyngeal kine receptor 1 in T-cell–mediated respiratory inflammation. Am
aspirates from wheezing children with respiratory syncytial virus J Pathol. 2008;172(2):386–394.
or influenza A virus infection. Pediatr Allergy Immunol. 2002; 72. Schelegle ES, Gershwin LJ, Miller LA, et al. Allergic asthma
13(5):350–356. induced in rhesus monkeys by house dust mite (Dermatopha-
57. Oriss TB, Ostroukhova M, Seguin-Devaux C, et al. Dynamics goides farinae). Am J Pathol. 2001;158(1):333–341.
of dendritic cell phenotype and interactions with CD4þ T cells 73. Schwarze J, Cieslewicz G, Hamelmann E, et al. IL-5 and eosino-
in airway inflammation and tolerance. J Immunol. 2005; phils are essential for the development of airway hyperrespon-
174(2):854–863. siveness following acute respiratory syncytial virus infection.
58. Papadopoulos NG, Moustaki M, Tsolia M, et al. Association of rhi- J Immunol. 1999;162(5):2997–3004.
novirus infection with increased disease severity in acute bronchio- 74. Schwarze J, Cieslewicz G, Joetham A, et al. Critical roles for
litis. Am J Respir Crit Care Med. 2002;165(9):1285–1289. interleukin-4 and interleukin-5 during respiratory syncytial virus
59. Parrott RH, Kim HW, Arrobio JO, et al. Epidemiology of respira- infection in the development of airway hyperresponsiveness after
tory syncytial virus infection in Washington, D.C., II: infection airway sensitization. Am J Respir Crit Care Med. 2000;162(2, pt
and disease with respect to age, immunologic status, race and sex. 1):380–386.
Am J Epidemiol. 1973;98(4):289–300. 75. Schwarze J, Hamelmann E, Bradley KL, et al. Respiratory syncy-
60. Peat JK, Tovey E, Toelle BG, et al. House dust mite allergens: a tial virus infection results in airway hyperresponsiveness and
major risk factor for childhood asthma in Australia. Am J Respir enhanced airway sensitization to allergen. J Clin Invest. 1997;
Crit Care Med. 1996;153(1):141–146. 100(1):226–233.
61. Peebles RS Jr, Sheller JR, Collins RD, et al. Respiratory syncytial 76. Shay DK, Holman RC, Newman RD, et al. Bronchiolitis-
virus infection does not increase allergen-induced type 2 cytokine associated hospitalizations among US children, 1980–1996.
production, yet increases airway hyperresponsiveness in mice. JAMA. 1999;282(15):1440–1446.
J Med Virol. 2001;63(2):178–188. 77. Shay DK, Holman RC, Roosevelt GE, et al. Bronchiolitis-
62. Peebles RS Jr, Sheller JR, Johnson JE, et al. Respiratory syncytial associated mortality and estimates of respiratory syncytial
virus infection prolongs methacholine-induced airway hyperre- virus–associated deaths among US children, 1979–1997. J Infect
sponsiveness in ovalbumin-sensitized mice. J Med Virol. 1999; Dis. 2001;183(1):16–22.
57(2):186–192. 78. Siegle JS, Hansbro N, Herbert C, et al. Early-life viral infection
63. Poulsen A, Stensballe LG, Nielsen J, et al. Long-term conse- and allergen exposure interact to induce an asthmatic phenotype
quences of respiratory syncytial virus acute lower respiratory tract in mice. Respir Res. 2010;11:14.
106 Veterinary Pathology 52(1)

79. Sigurs N, Aljassim F, Kjellman B, et al. Asthma and allergy pat- 90. Tasker RC, Gordon I, Kiff K. Time course of severe respiratory
terns over 18 years after severe RSV bronchiolitis in the first year syncytial virus infection in mechanically ventilated infants. Acta
of life. Thorax. 2010;65(12):1045–1052. Paediatr. 2000;89(8):938–941.
80. Sigurs N, Bjarnason R, Sigurbergsson F, et al. Respiratory syncy- 91. Taussig LM, Wright AL, Morgan WJ, et al. The Tucson Chil-
tial virus bronchiolitis in infancy is an important risk factor for dren’s Respiratory Study, I: design and implementation of a pro-
asthma and allergy at age 7. Am J Respir Crit Care Med. 2000; spective study of acute and chronic respiratory illness in
161(5):1501–1507. children. Am J Epidemiol. 1989;129(6):1219–1231.
81. Sigurs N, Gustafsson PM, Bjarnason R, et al. Severe respiratory 92. Techaarpornkul S, Barretto N, Peeples ME. Functional analysis
syncytial virus bronchiolitis in infancy and asthma and allergy of recombinant respiratory syncytial virus deletion mutants
at age 13. Am J Respir Crit Care Med. 2005;171(2):137–141. lacking the small hydrophobic and/or attachment glycoprotein
82. Simoes EA, Groothuis JR, Carbonell-Estrany X, et al. Palivizu- gene. J Virol. 2001;75(15):6825–6834.
mab prophylaxis, respiratory syncytial virus, and subsequent 93. Thompson WW, Shay DK, Weintraub E, et al. Mortality associ-
recurrent wheezing. J Pediatr. 2007;151(1):34–42, 42.e1. ated with influenza and respiratory syncytial virus in the United
83. Simoes EA, Hayward AR, Ponnuraj EM, et al. Respiratory syncy- States. JAMA. 2003;289(2):179–186.
tial virus infects the Bonnet monkey, Macaca radiata. Pediatr 94. Tian M, Liu F, Wen GY, et al. Effect of variation in RANTES
Dev Pathol. 1999;2(4):316–326. promoter on serum RANTES levels and risk of recurrent wheez-
84. Simpson BM, Custovic A, Simpson A, et al. NAC Manchester ing after RSV bronchiolitis in children from Han, southern
Asthma and Allergy Study (NACMAAS): risk factors for asthma China. Eur J Pediatr. 2009;168(8):963–967.
and allergic disorders in adults. Clin Exp Allergy. 2001;31(3): 95. Tremblay K, Lemire M, Provost V, et al. Association study
391–399. between the CX3CR1 gene and asthma. Genes Immun. 2006;
85. Sims DG, Downham MA, Gardner PS, et al. Study of 8-year-old 7(8):632–639.
children with a history of respiratory syncytial virus bronchiolitis 96. Tripp RA, Jones LP, Haynes LM, et al. CX3C chemokine mimi-
in infancy. Br Med J. 1978;1(6104):11–14. cry by respiratory syncytial virus G glycoprotein. Nat Immunol.
86. Sporik R, Holgate ST, Platts-Mills TA, et al. Exposure to house- 2001;2(8):732–738.
dust mite allergen (Der p I) and the development of asthma in 97. Wilson RH, Whitehead GS, Nakano H, et al. Allergic sensitiza-
childhood: a prospective study. N Engl J Med. 1990;323(8): tion through the airway primes Th17-dependent neutrophilia and
502–507. airway hyperresponsiveness. Am J Respir Crit Care Med. 2009;
87. Stein RT, Sherrill D, Morgan WJ, et al. Respiratory syncytial 180(8):720–730.
virus in early life and risk of wheeze and allergy by age 13 years. 98. Wu P, Dupont WD, Griffin MR, et al. Evidence of a causal role
Lancet. 1999;354(9178):541–545. of winter virus infection during infancy in early childhood
88. Stensballe LG, Simonsen JB, Thomsen SF, et al. The causal asthma. Am J Respir Crit Care Med. 2008;178(11):1123–1129.
direction in the association between respiratory syncytial virus 99. Yasue M, Nakamura S, Yokota T, et al. Experimental monkey
hospitalization and asthma. J Allergy Clin Immunol. 2009; model sensitized with mite antigen. Int Arch Allergy Immunol.
123(1):131–137.e131. 1998;115(4):303–311.
89. Stern DA, Guerra S, Halonen M, et al. Low IFN-gamma produc- 100. Young S, O’Keeffe PT, Arnott J, et al. Lung function, airway
tion in the first year of life as a predictor of wheeze during child- responsiveness, and respiratory symptoms before and after
hood. J Allergy Clin Immunol. 2007;120(4):835–841. bronchiolitis. Arch Dis Child. 1995;72(1):16–24.

You might also like