Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2004 Apr;17(2):390–412. doi: 10.1128/CMR.17.2.390-412.2004

Animal Pneumoviruses: Molecular Genetics and Pathogenesis

Andrew J Easton 1, Joseph B Domachowske 2, Helene F Rosenberg 3,*
PMCID: PMC387412  PMID: 15084507

Abstract

Pneumoviruses are single-stranded, negative-sense, nonsegmented RNA viruses of the family Paramyxoviridae, subfamily Pneumovirinae, and include pathogens that infect humans (respiratory syncytial virus and human metapneumovirus), domestic mammals (bovine, ovine, and caprine respiratory syncytial viruses), rodents (pneumonia virus of mice), and birds (avian metapneumovirus). Among the topics considered in this review are recent studies focused on the roles of the individual virus-encoded components in promoting virus replication as well as in altering and evading innate antiviral host defenses. Advances in the molecular technology of pneumoviruses and the emergence of recombinant pneumoviruses that are leading to improved virus-based vaccine formulations are also discussed. Since pneumovirus infection in natural hosts is associated with a profound inflammatory response that persists despite adequate antiviral therapy, we also review the recent experimental treatment strategies that have focused on combined antiviral, anti-inflammatory, and immunomodulatory approaches.

INTRODUCTION

Pneumoviruses (family Paramyxovirida, subfamily Pneumovirinae) are enveloped viruses with negative-sense, nonsegmented RNA genomes. The human pathogen, respiratory syncytial virus (hRSV) remains the best characterized of this group. Despite considerable efforts toward the development of effective treatments for hRSV disease, acute infection remains associated with significant morbidity and mortality, particularly among immunocompromised patients and infants born prematurely. Supportive therapy alone remains the standard of care for the treatment of severe cases of hRSV infection, although prophylactic treatments are now available and encouraging progress has been made toward formulating a vaccine. The recently discovered human pathogen human metapneumovirus (hMPV) also belongs to this virus subfamily, as do the veterinary pathogens avian metapneumovirus (APV), bovine respiratory syncytial virus (bRSV), ovine and caprine RSVs, and pneumonia virus of mice (PVM). The text begins with a review of pneumovirus biology and molecular genetics, continues with a discussion of current concepts related to the pathogenesis of pneumovirus disease as it occurs in vivo and as it is modeled in vitro, and concludes with a discussion of recent therapeutic options for the treatment of hRSV in human subjects.

MOLECULAR GENETICS

Virion Structure

Electron microscopic analysis of the virions of all four members of the subfamily Pneumovirinae demonstrates that they are structurally similar to one another and similar to other members of the family Paramyxoviridae. Virions, while pleomorphic, are typically spherical (diameters of 150 to 200 nm), although filamentous particles of up to 400 nm in length have been described (9, 22, 192). PVM virions are predominantly filamentous, 100 to 120 nm in diameter, and up to 3 μm in length (71). The functional differences (if any) based on virion structure are not known, and the differences observed may be a consequence of the cell type used for infection. A diagrammatic representation of a typical pneumovirus is shown in Fig. 1. The pneumovirus virion is surrounded by a lipid envelope derived from the plasma membrane of the host cell into which the three virus glycoproteins, the attachment (G), fusion (F), and small hydrophobic (SH) proteins, are inserted (59, 176). The F protein spans the lipid membrane, with the amino terminus located outside the virion and a short cytoplasmic carboxy-terminal “tail” located inside. In contrast, the G protein is a type 2 membrane protein, with internal amino and external carboxy-terminal components. The F and G proteins comprise the 10- to 14-nm spikes on the virion surface (71). The precise localization of the SH glycoprotein has not yet been established.

FIG. 1.

FIG. 1.

Schematic of the pneumovirus particle. The RNA genome associates with viral proteins to form the helical nucleocapsid structure (represented on the right and in the center of the virion on the left). The proteins consist of the nucleocapsid protein (N), the phosphoprotein (P), and the large polymerase (L) protein. The M2-1 protein is also thought to be present in this complex (not shown). The nucleocapsid structure is surrounded by the matrix (M) protein, which forms a link between the nucleocapsid and the lipid membrane of the virus particle. Embedded in the lipid membrane are the attachment (G) glycoprotein, the fusion (F) protein, and the small hydrophobic (SH) protein (not shown).

The F and G glycoproteins interact with the virus matrix (M) protein layer on the internal surface of the membrane. The details of the molecular interaction(s) between the M protein and the surface F and G glycoprotein(s) are not clear.

The helical nucleocapsid is located within the M-protein layer, and includes the 13- to 15-kb single-stranded, non-segmented RNA genome in association with the virus nucleoprotein (N) and large (L) proteins and the phosphoprotein (P). The pneumovirus nucleocapsids (diameter, 13 to 14 nm) are significantly smaller than those that have been described for other paramyxoviruses (18 nm) (22). Electron microscopic studies indicate that hRSV nucleocapsids are less rigid and narrower than those of measles virus and Simian virus 5 (SV5), further confirming differences in nucleocapsid morphology between the Pneumovirinae and the Paramyxovirinae (23). The L protein is often referred to as the RNA polymerase since it is thought to contain all of the catalytic activities necessary for virus RNA synthesis, although it does not function as such without the N and P proteins. The M2-1 transcriptional enhancer protein is also thought to be associated with the nucleocapsid, although this has not yet been determined directly.

Virus Attachment and Entry

The conventional model for pneumovirus attachment involves the virus G protein interacting with a molecule or molecules on the target cell surface (232). While the precise nature of the cellular receptor has not been identified, current evidence suggests that one or more cellular glycosaminoglycans or heparin-like molecules are involved (116, 163, 164, 218, 342). Following G-protein-mediated attachment, the model proposes that the F glycoprotein then promotes pH-independent fusion between the cell membrane and the virus envelope via a mechanism involving the hydrophobic amino-terminal region of the F1 component. The fusion process introduces the internal components of the virion into the cytoplasm of the host cell, where the remainder of the infectious cycle takes place. While pneumovirus infection results in alteration of host cell gene expression, there is no absolute requirement for a functional nucleus, since virus development can proceed in enucleated cells (122).

Recent analyses of deletion mutants of RSV have raised questions about the conventional model of attachment. Karron et al. (205) have characterized a mutant hRSV strain, cp-52, that grows in tissue culture but is attenuated in animals and humans. Sequencing of the genome of this mutant showed that it was lacking both the G and SH coding regions but retained the F-protein gene. Using a recombinant vaccinia virus system. Heminway et al. (172) suggested that the hRSV fusion event required the presence of the F, G, and SH proteins acting in concert. However, Kahn et al. (199) demonstrated that heterologous expression of the hRSV F protein alone promoted infection and cell fusion. Several mutants which lack either the G gene or the SH gene or both have been generated using a reverse genetics approach (341, 342; see below). All of these mutants are viable in tissue culture; their growth rates vary depending on the target cell and are significantly attenuated in vivo compared to that of the wild-type parental strain. There are also data indicating a binding interaction between the RSV G protein and the fractalkine receptor, CX3CR1 (353), although the latter is not expressed on human epithelial cells. These data suggest that the attachment and fusion steps may be more complex than the conventional model proposes.

Genome Organization of the Pneumovirinae

The genomes of the pneumoviruses are ∼15,000 nucleotides in length (262, 332; L. Thorpe and A. Easton, unpublished data). Metapneumovirus genomes are smaller (∼13,000 kb) than those of the pneumoviruses. While the precise sizes of the genomes are strain dependent, the number of nucleotides does not follow the “rule of six” (42, 43, 312), a feature that differentiates the Pneumovirinae from the Paramyxovirinae. The reasons underlying the discrepancy have not been determined, but it has been proposed that length restriction among the Paramyxovirinae may be due to specific N-protein to RNA interactions (e.g., binding units of six nucleotides) and/or the need to ensure the integrity of the genome in viruses susceptible to non-template-dependent nucleotide insertion.

Figure 2 summarizes the genome organization of the pneumoviruses and metapneumoviruses together with representatives of the Paramyxovirinae. While the general strategy for genome organization is similar for all of the viruses, there are differences in specific detail. The genomes of the pneumoviruses and metapneumoviruses encode a greater number of mRNAs (10 and 8, respectively) than the 6 or 7 mRNAs encoded by genomes of the Paramyxovirinae. While all members of the family Paramyxoviridae encode a standard set of structural proteins (293), one feature which differentiates the Paramyxovirinae from the Pneumovirinae are different coding potentials of P genes and the use of an RNA editing mechanism to express multiple proteins from this gene (82). Furthermore, the Pneumovirinae lack counterparts of the C and V proteins found in Paramyxovirinae, although the PVM P gene does direct the synthesis of a polypeptide by using a second open reading frame in a manner analogous to the expression of the paramyxovirus C proteins (14). Another unique feature of the Pneumovirinae is the gene encoding the M2 proteins which have not been identified in any other virus genome.

FIG. 2.

FIG. 2.

Genomic structures of viruses of the family Paramyxoviridae. The subfamilies, genera, and representative viruses are as indicated. *, pneumonia virus of mice is similar, without the M2-L gene overlap.

The genomes of the Pneumovirinae contain regulatory sequences at each terminus. The 3′ end of the genome sense RNA directs both replication and transcription, while the 5′ end of the genome RNA (i.e., the 3′ end of the antigenome) contains signals that direct replication of the antigenome for synthesis of progeny virions. The metapneumovirus genome encodes eight distinct transcription units in a linear array, with each unit separated by a short segment of untranscribed sequence. In contrast, the pneumovirus genome encodes 10 transcription units, including nonstructural proteins NS1 and NS2 (106). There is one notable difference in gene order: the F-protein gene follows the G-protein gene in the pneumovirus genomes, the reverse order from that observed among the metapneumoviruses (293). In evolutionary terms, this difference is very striking since recombination among these virus genomes has been observed only in the laboratory setting (329).

While the PVM genome is organized with a tandem array of distinct genes similar to that of the metapneumoviruses, the hRSV genome includes a 68-nucleotide overlap of the gene encoding the M2 proteins with the start of the L gene, discussed further in “Current model for genome transcription” (below).

Pneumovirus Gene Products

As indicated in Fig. 2, the nucleocapsid, phosphoprotein, matrix, fusion, and polymerase proteins are common to all members of the family Paramyxoviridae. Much of our understanding of the functions of these proteins has been derived by extrapolation from experiments performed with one or more specific viruses, particularly Sendai virus. More recently, it has been possible to confirm these functions directly by targeted mutagenesis and reverse genetics.

Nonstructural (NS1 and NS2) proteins.

The NS1 and NS2 proteins are unique to the pneumoviruses. The genes are located at the 3′-most positions of the genome and, as a consequence, are the most abundantly transcribed genes. The NS1 and NS2 proteins are relatively small, with the NS1 proteins of hRSV and PVM being 139 and 113 amino acids in length, respectively, and the NS2 proteins being 124 and 156 amino acids, respectively (50, 57, 58).

The hRSV NS1 protein interacts with the M protein in infected cells, and two-hybrid analysis identified an additional interaction between NS1 and the P protein (106, 173). In reverse genetics studies, expression of the NS proteins inhibited RNA synthesis, suggesting a regulatory role for the NS proteins in vivo (8, 343). While the finding of hRSV mutants lacking the NS1 or NS2 genes confirmed that the NS proteins are dispensable for virus replication in vitro, recombinant viruses grew more poorly than the parental virus and the mutants were attenuated in chimpanzees (188, 344, 347, 378).

Other studies suggest that the pneumovirus NS proteins play a role in inhibiting responses to interferon (30, 31, 315). Recombinant rabies viruses each expressing one of the bovine RSV NS genes were resistant to interferon treatment if cells were dually infected with the virus but not if they were infected alone (315), suggesting that the NS1 and NS2 proteins function together to inhibit responses to interferon in this setting. Most recently, Bossert et al. (31) have demonstrated that NS1 and NS2 proteins inhibit the activation of interferon regulatory factor 3 and thus the production of beta interferon in bRSV- infected MDBK cells. Valarcher et al. (358) likewise demonstrated that recombinant, NS-2 lacking, bRSV are potent inducers of interferon in bovine nasal fibroblasts and bronchoalveolar macrophages. Studies using recombinant rabies viruses that express the PVM or hRSV NS1 and NS2 genes have yielded similar results (30, B. Bossert, A. J. Easton, and K. K. Conzelmann, unpublished data). The mechanism by which the interferon inhibition is achieved is not yet understood.

Nucleocapsid (N) protein.

The N protein forms an integral part of the nucleocapsid complex of the virion and is an essential component of the polymerase complex. The N protein is thought to be responsible for giving the RNA genome its helical structure through a tight association with the virus genome RNA; interestingly, hRSV N protein expressed in insect cells spontaneously formed nucleocapsid structures containing RNA (24). While the RSV N protein (391 amino acids) is smaller than its counterparts among the Paramyxovirinae (489 to 553 amino acids) and shows little sequence similarity, alignments with the amino-terminal 400 amino acids of the N proteins of various nonsegmented negative-sense RNA viruses suggested similarities among predicted secondary structures (13). The hRSV N protein has a relatively high level of amino acid identity (60%) to the N protein of PVM (13) and has regions of specific sequence homology to the N proteins of APV and hMPV (234, 361).

Deletion mutant analysis of N protein expressed in cells suggests that a large segment of this protein is required for interacting with the P protein (128). Barr and Easton (12) demonstrated that the amino-terminal half of the PVM N protein was unable to bind to immobilized P protein while the carboxy-terminal half of the N protein bound with only 17.6% of the affinity seen with the full-length protein, results suggesting that the complete N protein is important for thisinteraction.

Phosphoprotein (P).

The P protein is an essential component of the replication and transcription complexes of the pneumoviruses (14, 225, 234, 314, 361). Analysis of the interaction between the P and N proteins has shown that the carboxy terminus of the P protein contains most of the elements necessary to bind to the N protein (128, 173, 207, 242, 247, 326). The P-protein genes of the Pneumovirinae encode several different proteins using alternative start codons. In addition to the full-length sequence, the P gene of hRSV encodes an mRNA for an additional, short polypeptide of unknown function (45). While the PVM and hRSV P proteins have significant amino acid identity, the two proteins align well only at the extreme amino terminus and within the carboxy-terminal half of the protein, and there are large regions near the amino terminus with limited to nonexistent sequence identity (14), including the second open reading frame of the PVM P gene. The protein product of this second open reading frame has been detected in infected cells, as have four additional proteins generated by initiation at internal AUG translation initiation codons within the P-protein gene (14). No additional proteins have been identified from the P genes of the meta-pneumoviruses.

The P protein is phosphorylated at specific serine residues (98). Villanueva et al. (366) demonstrated that most of the phosphorylation, occurring at residues 116, 117, 119, and 232, is not essential for transcription or replication.

Matrix (M) protein.

The M protein is associated with the inner face of the lipid membrane of infected target cells (284, 382). All M proteins include a carboxy-terminal hydrophobic domain. The M proteins of nonsegmented minus-strand viruses are thought to function by rendering the nucleocapsid transcriptionally inactive before packaging and also by promoting the association of the nucleocapsid complex with the nascent envelope (140, 343).

Small hydrophobic (SH) protein.

The SH proteins of pneumoviruses are small integral membrane proteins. The sizes of the proteins vary considerably, with hRSV having the smallest SH protein (64 amino acids) and hMPV having the largest (183 amino acids) (58, 100, 361). The RSV SH protein is anchored in the membrane by a hydrophobic signal-anchor sequence (275) and is a type 2 membrane protein (59, 117). The precise function of the SH protein is unknown, and analysis of hRSV mutants lacking the SH and/or G genes suggests that the SH protein is not absolutely necessary for attachment, infectivity, or virion assembly. However, the hRSV G and SH proteins have also been implicated in impairing the Th1-mediated host antiviral responses (352).

Attachment (G) protein.

The hRSV G protein was identified as the viral attachment protein based on the observation that G-specific polyclonal antibody blocked absorption of the virus to the surface of target cells (232). Similarly, a monoclonal antibody that inhibited the hemagglutinating activity of PVM likewise bound to the G protein (235). The pneumovirus G proteins do not show any sequence or structural homology to the HN or H attachment proteins of other viruses of the family Paramyxoviridae, although all are type 2 glycoproteins (373). The G proteins of pneumoviruses are heavily glycosylated with both N- and O-glycosylated sites, adding approximately 55 kDa to the mass of the hRSV polypeptide. While the roles of these glycosyl groups have not been completely clarified, extensive glycosylation of the extracellular domain of the G protein could reduce its antigenicity by shielding the virus protein with host-specified sugars. Alternatively, partial utilization of glycosylation sites can result in antigenic heterogeneity, representing a source of diversity within a genetically homogenous virus population. The immune response to hRSV correlates with and provides selection pressure for G-protein variation (127, 261).

The hRSV G protein in infected cells is present in two forms. The first is translated from the entire open reading frame and contains an amino-terminal sequence prior to the putative signal/membrane anchor region. A second, secreted form of the protein has been observed (304) which is generated by the initiation of translation at an internal AUG initiation codon that is in the same reading frame as that used for the full- length G protein. The function of the secreted protein is not known, but it may alter the immune response of the host, possibly by acting as a “decoy” or by altering specific aspects of the host inflammatory response (190, 191).

While the G protein of PVM shares structural features with those of hRSV and the metapneumoviruses (295), the G-protein genes of two strains of PVM have been sequenced and show intriguing differences. The G protein of the nonpathogenic PVM strain 15 does not have an amino-terminal sequence prior to the putative membrane anchor, while the G gene of a pathogenic PVM strain contains a single base deletion and several mutations (295).

A final interesting feature of the G genes of hRSV and PVM is the presence of a small open reading frame just 5′ to the main coding region, of function unknown (295, 373).

Fusion (F) protein.

The F protein of hRSV was first was identified by immunoprecipitation using monoclonal antibodies which inhibit the formation of multicell syncytia in cell culture. Although the pneumovirus F proteins show only limited amino acid sequence identity to the F proteins of other viruses of the family Paramyxoviridae, they show significant structural similarities to members of this group (15, 18, 51, 64, 361, 386). The F protein of hRSV is synthesized as an inactive precursor (F0) that assembles into a homo-oligomer in the rough endoplasmic reticulum (ER), forming a structure that is presumed to represent a single virion spike (62). When the F0 protein reaches the trans Golgi network, it is activated by cleavage into two disulfide-linked subunits, F1 and F2, at two furin consensus sites (142, 392, 393). The short (27-amino-acid) peptide between the two furin cleavage sites does not remain associated with the F-protein subunits (19) and a novel biological activity for this protein has recently been described (314; see “Virus evasion of innate immunity” below).

M2 proteins.

The genes encoding the M2 proteins of pneumoviruses and metapneumoviruses contain two overlapping open reading frames (Fig. 3) (4, 58, 165, 237, 361, 387). The first open reading frame, initiated by the 5′-proximal AUG initiation codon, encodes the M2-1 protein, which is involved in virus RNA synthesis as described below. Immunocytochemical analysis of cytoplasmic inclusions showed that genome RNA, N, P, L, and M2-1 proteins were colocalized in hRSV-infected cells (126). Reverse genetics studies have shown that while the minimal trans-acting requirements for hRSV (i.e., proteins that must be added for efficient first-round virus replication) are the N, P, and L proteins, the presence of the M2 gene significantly enhanced the production of full length virus mRNA (62, 63, 112, 152, 388). In cells expressing the N, P, and L proteins, virus-specific transcription resulted in the production of prematurely terminated mRNA (62) and expression of the M2-1 protein served to promote the production of full- length mRNA without altering genome RNA replication (63). These data suggest that the M2-1 protein functions as a transcription elongation factor.

FIG. 3.

FIG. 3.

Diagrammatic representation of the organization of the pneumovirus M2-1 and M2-2 genes. The grey bar represents the M2-1 open reading frame, and the white bar represents the M2-2 open reading frame. The positions of the translation initiation and termination codons of are identified. aa, amino acids.

The product of the second open reading frame, the M2-2 protein, has been detected in cells infected with hRSV and PVM (4). No M2-2 protein was detected in APV-infected cells, and to date there is no information about the putative M2-2 protein of hMPV. Expression of the M2-2 open reading frame resulted in the inhibition of virus gene expression from synthetic genomes. The RSV M2-2 protein accumulates during infection, and it has been proposed that the role of this protein is to switch the virus from replicative to assembly mode prior to virion release (4, 22).

Large (L) polymerase protein.

By analogy to other viruses, the pneumovirus L protein is thought to be the major component of the viral RNA-dependent RNA polymerase complex, which is responsible for the synthesis of all viral RNA, including mRNA, replicative intermediates, and the progeny RNA genomes. The L protein is also thought to be responsible for mRNA methylation and capping, although this has not been shown directly. As the name indicates, the L protein is very large—the hRSV L protein contains 2,165 amino acids, and the hMPV L protein contains 2,000 amino acids (297, 361). Poch et al. (289, 290) described six conserved domains for polymerase proteins of minus-strand RNA viruses, including three functional domains and distinct interdomain regions acting as hinges to assemble the functional domains into an appropriate conformation. Domain III contains the polymerase GDNQ motif, and domain IV contains a putative ATP binding site (200). There have been few direct studies of the specifics of the L proteins of the Pneumovirinae. Most recently, Cartee et al. (47) demonstrated that a single amino acid change (N1049D) resulted in aberrant transcriptional termination without altering the rate of virus replication.

Current Model for Transcription

The details of the current model of transcription and replication of the pneumovirus genomes are consistent with those proposed for all nonsegmented negative-sense RNA viruses. The virus genome contains a single polymerase entry site, termed the leader or promoter sequence, at the 3′ end of the RNA. The polymerase complex initiates transcription at the conserved transcriptional start sequence that is present at the beginning of all transcribed genes. The polymerase then travels along the genome until it reaches a conserved signal that marks the point at which transcription terminates. At some point during the transcription process, the nascent mRNA is methylated to produce a cap structure that ensures translation on host cell ribosomes; there are no reports directly addressing this process. The signal marking the end of transcription among the pneumoviruses contains a short run of U residues, and this marks the position at which the polyadenylated tail is added to the mRNA. When the polyadenylation process is complete, the mRNA is released for translation on either free or membrane-bound ribosomes, depending on the nature of the protein product. At this point, the polymerase does not automatically progress along the genome. Rather, only a proportion of the active polymerase molecules do so, with estimates suggesting that approximately 50% of the polymerase molecules are able to continue. The dissociated polymerases can reinitiate transcription only by translocation to the 3′-terminal leader region to begin transcription at the first gene. The remaining polymerases traverse the genome without transcribing until they encounter the conserved transcriptional start sequence of the second gene. At the end of this gene, the remaining polymerases repeat the dissociation-versus-continuation process. The result is that genes proximal to the 3′ leader region are represented in mRNA more abundantly than are those closer to the 5′ end in a gradient of transcription (215). The intergenic regions of the pneumoviruses are variable in length and tolerate mutation and insertion of additional nucleotides (18, 42, 49, 361).

The process of termination and reinitiation is poorly understood, but the M2-1 protein of RSV has been shown to be essential for production of full-length virus mRNA (62, 63, 112, 166). The antitermination activity during transcription permits the viral polymerase to remain associated with the template, thereby increasing the production of full-length mRNA and enhancing the production of products of genes that are distal to the 3′ leader region. The eight RSV gene junctions, which vary in their ability to terminate transcription, all direct the production of more readthrough mRNA in the presence of M2-1 protein, although they vary in their sensitivity to the presence of the M2-1 protein (167).

The genome structure of hRSV contains an exception to the process described above. The transcriptional stop signal for the M2 gene is located 68 nucleotides downstream of the transcriptional start sequence for the L gene (65). Thus, a large proportion of the mRNA initiated at the L-gene start sequence terminates at the M2 transcriptional stop sequence (65, 111).

Elements within intergenic regions play a vital role in the regulation of transcription (114). The intergenic regions of the Pneumovirinae show considerable sequence diversity and vary from 2 to 56 nucleotides in length. The transcriptional start sequences are thought to be 10 nucleotides in length, and the consensus sequences, while conserved, are slightly different for each pneumovirus (61, 221). The PVM transcriptional start signals are more variable than those of RSV or APV, which are absolutely conserved with the exception of the L gene (49, 296, 332).

The transcriptional stop signals of hRSV consist of a conserved pentanucleotide sequence together with a 1- to 4-nucleotide AU-rich region and a 4- to 7-nucleotide poly(U) tract. Saturation mutagenesis of the hRSV gene transcriptional start sequences showed that residues 1, 3, 6, 7, and 9 were critical in directing transcription initiation, although there was some variability in efficiency, particularly with the NS1 and NS2 gene start sequences, which were approximately 40% less efficient than the others (219, 220, 221).

Deletion of a transcriptional stop signal from an upstream gene resulted in transcriptional readthrough to the next gene without termination. Harmon et al. (168) analyzed the hRSV M-gene transcriptional stop sequence and determined that the integrity of the pentanucleotide and AU-rich regions was essential for efficient termination. The residue following the poly(U) tract might also be important for efficient transcription termination (335).

No significant difference in transcriptional activity or efficiency was seen with different sized RSV intergenic sequences (167, 219). Bukreyev et al. (42) described the recovery of a recombinant hRSV containing an intergenic region of 160 nucleotides. This very long intergenic region had little effect on sequential transcription.

The genome structure of hRSV contains an exception to the process described above. The gene end signal for the M2 gene is located 68 nucleotides downstream of the gene start sequence for the L gene (65), and much of the mRNA that initiates at the L transcriptional start site terminates at the M2 gene end sequence (65, 111). The role (if any) of this truncated L transcript remains to be elucidated.

Current Model for Genome Replication

The current model, based on that determined for other nonsegmented negative-sense RNA viruses, proposes that the nucleocapsid complex is the functional unit for both replication and transcription and that the L, N, and P proteins are involved in both processes. Transcription and replication promoters of hRSV overlap but are not identical (114). In the replicative mode, the polymerase binds to the 3′ end of the genome RNA and initiates RNA synthesis de novo. Once initiated, the polymerase is committed to continue to the end of the template to produce an antigenome. The polymerase complex then uses the antigenome as a template, binding to and initiating RNA synthesis at the 3′ end of the antigenome RNA. In the genome sense RNA, the trailer region contains the antisense copy of the sequence necessary for initiating replication to produce more genome RNA from the antigenome.

It is not yet clear precisely how the polymerase complex alters its activity and switches from the transcription to the replication mode. Fearns et al. (113) showed that increased expression of the N protein stimulated hRSV replication. This is consistent with the proposal made for other negative-sense RNA viruses, suggesting that replication is dependent on RNA encapsidation (224). However, there are some conflicting data suggesting that high levels of N protein may also stimulate transcription, and, more recently, a role for M2-2 protein has been proposed (21).

The 3′ and 5′ termini of the pneumovirus genome contain the sequences that direct replication. Reverse genetics studies showed that the 3′-terminal 44-nucleotide leader region and the 5′-terminal 40 nucleotides of the trailer region of hRSV were necessary for replication, encapsidation, and assembly (66). The immediate termini of the pneumovirus genomes have complementary sequences, as has been described for other negative-sense RNA viruses (262, 296, 361). The trailer sequences are more efficient than the leader sequences at directing replication, as would be expected from their role in replication and the requirement for the leader region to direct both replication and transcription (114).

Analysis of the leader and trailer sequences suggested that nucleotide positions 1, 2, 3, 5, 6, and 7 were particularly important for replication, while position 4 was found to be tolerant of alteration (285). Using synthetic chimeric minigenomes with terminal sequences from both APV and RSV in virus infected cells, Marriott et al. (250) showed that paired termini from the same virus were required for replication to occur. It is not clear whether the critical step is replication or encapsidation, but the data suggest that an association between the leader and trailer is necessary for productive infection (250).

Packaging and Assembly

Reverse genetics experiments suggest that the accumulation of the M protein may be a component of the trigger to initiate the assembly of virions (343). It is anticipated that the nucleocapsid complex/M-protein structure associates with the internal tails of the major glycoproteins that are inserted into the membrane of the infected cell after processing through the Golgi complex. In parallel with studies performed with other viruses, a progressive series of interactions between the glycoproteins and the M protein, which acts as a bridge with the nucleocapsids, results in the progeny virions budding from the surface of the cell in the form of filament structures (280). In polarized cells, such as those of the respiratory tract, hRSV buds from the apical surface and is shed into the airways, from which it can be transmitted to new hosts (303).

PATHOGENESIS OF DISEASE

This section is divided into four parts. APV, hMPV, and bRSV infections are discussed individually. In the fourth part, we consider the human pneumovirus pathogen hRSV. The current state of understanding of the human disease as assessed by direct studies of patients and patient-derived materials is reviewed, followed by the results obtained from cell culture and animal models of hRSV. Consideration of the mouse pneumovirus pathogen mPVM has been interwoven with the information about hRSV, since it has been developed specifically as a surrogate for severe hRSV infection in rodent models.

Avian Metapneumovirus Infection

APV, a metapneumovirus previously known as turkey rhinotracheitis virus, is the cause of severe respiratory infection in turkeys (reviewed in references 73, 244 and 274) and may also be the etiologic agent of swollen head syndrome in chickens (73). Originally identified in South Africa in the late 1970s, APV was later found in Europe, Asia, and South America and has only recently emerged among flocks of domestic birds in the United States. Sequence divergence among the genes encoding the APV viral G (attachment) proteins suggested division of the European isolates into two subtypes, APV/A (in the United Kingdom) and APV/B (in continental Europe). A new subtype, APV/C, first emerged in the United States in 1996 in Colorado and was followed by a 1997 outbreak in Minnesota. Sequence analysis of APV/C isolates indicates that it is the most closely related to the newly discovered human metapneumovirus pathogen, hMPV (5, 274, 360). The U.S. outbreaks of APV infection have occurred in a seasonal pattern (spring and autumn), and several studies have implicated wild migratory birds in the spread of the disease (321). This disease has had devastating consequences for the turkey industry in Minnesota, resulting in much recent attention to disease detection, pathogenesis, and prevention (74, 322).

In the wild, APV leads to severe upper respiratory infection, often resulting in death of the infected bird. The clinical syndrome and the pathology observed in response to experimental infection with APV/A, APV/B, and APV/C (the last of these from the Colorado outbreak) have been described in several reports (75, 76, 193, 194, 277), in addition to a recent experimental study describing the natural history of APV/C from Minnesota (189). In the natural history study, respiratory symptoms were detected as early as 2 days postinoculation and included open mouth breathing, sneezing, swollen sinuses, and nasal discharge. Microscopically, inflammatory infiltrates (lymphocytes, macrophages, plasma cells, and heterophils) were detected in nasal turbinate and sinus tissues, although, interestingly, the ciliated epithelium, which was subjected to significant disruption in the earlier studies (including that describing APV/C from Colorado), remained intact. Anti-APV antibody responses emerged at 7 days postinoculation, although it is unclear whether this antibody response is protective (271). While the events promoting disease and tissue destruction are not yet understood at the molecular level, Munir and Kapur (266) have recently presented the results of a subtractive hybridization and gene microarray approach and have reported increased expression of transcripts encoding interferon response proteins and other elements of the inflammatory response and ubiquitination pathways in cultured chicken embryo cells infected with APV/C.

As might be anticipated, significant research has gone into vaccine studies. In various trials, several groups have demonstrated protection in response to immunization with high-passage virus (154, 283, 363, 381), with one report using high-passage virus from the Minnesota outbreak (APV/MN/turkey/1-a/97) together with the immunomodulatory agent S-28828 (299), an imidazoquinolinamine agent that induces interferon production and macrophage activation in avian species (203).

Human Metapneumovirus Infection

The very recently discovered hMPV is the first human pathogen of this family to be identified and is most closely related to the APVs described above; evolutionary analysis suggests that hMPV has existed in the human population for at least 50 years. This virus was first identified in the Netherlands by van den Hoogen et al. (360), and evidence of infection has since been collected worldwide (26, 105, 110, 273, 286), with one reported fatality in a stem cell transplant recipient (44). The clinical syndrome is currently indistinguishable from that resulting from hRSV infection, with some cases characterized by upper respiratory tract infection and others characterized by severe bronchiolitis and pneumonia (123). While several reports have associated hMPV infection with asthma exacerbation (123, 187, 247, 337), one report suggests that the association is less strong than that of asthma and rhinovirus infection (300). The current diagnostic approach involves reverse transcriptase PCR (RT-PCR) (245), facilitated by the substantial amount of virus gene sequence that is available (18, 245, 360, 361). Similar to what has been reported for hRSV infection, one group has reported acute wheezing and elevated concentrations of interleukin-8 (IL-8) in nasal secretions from patients infected with hMPV (187). For more detail, the reader is referred to two excellent recent reviews on this subject (197, 198).

Why was this virus invisible prior to 2001? The authors of the original study (360) discuss some possibilities, including very slow replication of hMPV in the standard cell lines used to isolate respiratory virus pathogens. Among the other possibilities, most clinical cases of hMPV disease are not life-threatening, and, as such, the impetus to identify novel pathogens among the pool of hRSV-negative respiratory infections has not been overwhelming. One can only wonder what other pathogens have been overlooked.

Bovine Pneumovirus Infection

Research on bRSV has focused primarily on issues related to its prevalence as a respiratory virus pathogen in dairy and beef cattle worldwide (reviewed in references 10, 103, 227, and 355). There are also distinct ovine and caprine RSV pathogens, infecting sheep and goats, respectively (reviewed in references 34 and 97). bRSV was first isolated and identified as a unique pathogen in the 1970s, and it is currently the closest known phylogenetic relative of hRSV (360), although there is no evidence for antigenic grouping into A/B strains (104). Similar to hRSV, bRSV results in seasonal infections among domestic cattle herds, and transmission is thought to occur via contact with respiratory secretions, with a short incubation period followed by signs and symptoms of upper and lower respiratory tract infection. Interestingly, and unlike hRSV infection, natural bRSV infection is often accompanied by concomitant bacterial infection (Mannheimia haemolytica, Pasteurella multocida, and Haemophilus somnus), resulting in what has been defined as the bovine respiratory disease complex. One study has documented sequence clustering based on geography (357), while yet another reports extensive sequence divergence in outbreaks distributed over time within a single herd (228).

There are several vaccine formulations in existence—modified live and inactivated, in both adjuvanted and nonadjuvanted forms (102, 374, 375)—and there is significant debate about the relative efficacy and modes of appropriate response. Modified live vaccines preferentially stimulate the production of neutralizing antibodies that detect the virus F (fusion) protein and stimulate lymphocyte proliferation and cytokine responses. Inactivated vaccines also stimulate lymphocyte proliferation and production of virus neutralizing antibodies, but the latter are produced together with nonneutralizing antibodies. There is some information relating to a potential immunopotentiation in response to challenge after formalin-inactivated vaccine administration (138, 318), analogous to what was observed in the human trials. While much of the focus has been on the development of humoral immunity to bRSV, recent results suggest that vaccine efficacy may relate to the development of cellular immunity, specifically Th1 (gamma interferon) responses (102, 121, 125, 374).

Experimental models of bRSV infection in calves involve aerosol administration of virions, resulting in early-onset cough and nasal discharge and progressing to signs and symptoms of lower respiratory tract infection (287, 350, 367, 380). In the study performed by Philippou et al. (287), lungs from animals sacrificed at 12 weeks postinoculation were atelectatic with evidence of bronchiolar obstruction and profound inflammatory infiltration. Disease progression has been associated with increased expression of IL-2, IL-4, and/or gamma interferon in cells drained from pulmonary lymph (139, 214) and in peripheral blood mononuclear cells (257), detection of tumor necrosis factor alpha and leukotriene B4 in respiratory secretions (302, 308), and elevations in levels of serum acute-phase response proteins (171). Aside from the challenging aspects of working with large experimental animals, research in this field is hampered by the fact that there are comparatively few bovine cytokine gene sequences available in the public domain and even fewer commercially available reagents.

Buchholz et al. (35) described the creation of recombinant bRSV virions in vitro utilizing the complete sequence genome of bRSV (389) by methods analogous to those developed for the study of hRSV. Among the results as they relate to disease pathogenesis are the finding that the bRSV NS1 and NS2 proteins function in synchrony to antagonize alpha/beta interferon-mediated antiviral activities in a species-specific fashion (30, 315) and that the SH and G genes are dispensable for bRSV replication in vitro (204). This technology has already been applied to the development of the next generation of bRSV and hRSV vaccines (36, 317).

Human Respiratory Syncytial Virus Infection

Human RSV is a seasonal infection, which in North America is observed most frequently in winter and spring. Transmission between individuals occurs via contact with respiratory secretions, either directly or via handled surfaces and objects or via large aerosolized droplets. The incubation period is typically 2 to 8 days, with virus replication spreading from the upper respiratory tract epithelial cells to the lower respiratory tract. The clinical signs and symptoms of acute hRSV infection have been described in significant detail in several recent reviews (86, 141, 155, 161, 162, 216, 372). The complex interplay of factors involved in acquired immunity to hRSV is discussed in the section on vaccines. The focus here is on several specific topics of recent interest.

Genetic susceptibility to severe infection.

While hRSV infection appears to be universal, there is great interest in understanding why and how some children proceed from initial infection to severe, life-threatening forms of lower respiratory tract disease. There are several straightforward and clearly understood conditions that lead to increased susceptibility to severe infection, including premature birth, congenital dysplasias, and immunodeficiency disorders (reviewed in references 52, 96, 260, 340, and 371). Less well understood are the conditions under which otherwise “normal” children succumb to severe hRSV disease. Several groups have approached this question via genotyping and have focused on questions relating to normal variation within the human population. Among the results, Hallman and colleagues (223, 239) have documented correlations between specific alleles of the genes encoding surfactant A and D with increased disease severity and Kwiatkowski and colleagues (177, 178) have provided evidence supporting a relationship between disease and a susceptibility locus related to the gene encoding IL-8. Two groups, Choi et al. (56) and Hoebee et al. (175), have identified alleles of IL-4, and IL-4 and its receptor, respectively, that are also associated with more severe sequelae of RSV infection in vivo. At present, it is not clear how these genetic factors influence responses to hRSV infection at the molecular level.

Inflammatory responses to infection.

To understand the concepts underlying inflammatory responses to hRSV, it is important to recognize the there are two different forms of hRSV-related disease discussed in the literature. Primary hRSV infection, or natural hRSV infection, occurs in a naive (i.e., unvaccinated) host, and the inflammatory responses are those of the innate immune system responding to virus replication in situ. This is to be contrasted with what has been termed “enhanced” or “augmented” disease, which describes the aberrant inflammatory responses observed in children who had been vaccinated with formalin-fixed virus preparations and later challenged “in the wild” with natural virus infection. The postvaccination response has been characterized extensively, and the profound lymphocytic and eosinophilic inflammation observed has been attributed to an induced Th1-Th2 imbalance, with dominance of Th2 lymphocyte responses in this setting (32, 148, 180).

With respect to primary hRSV infection, the earliest histologic studies were, of necessity, examinations of postmortem specimens. Severe infection progressed to include necrosis of airway epithelium, interstitial inflammation, and mucus secretion leading to airway obstruction and respiratory compromise. While lymphocytes were predominant in postmortem specimens, more recent studies have pointed to neutrophil influx as an earlier acute cellular response and to neutrophil-mediated activities as a major cause of respiratory symptoms (107, 209, 328, 370). Neutrophils are proinflammatory leukocytes of the granulocyte series that develop from pluripotent stem cells in the bone marrow, released into the circulation, and recruited into tissues via the coordinated actions of various cytokines and chemokines. Activated neutrophils participate in phagocytosis and degranulation and release reactive oxygen metabolites into the surrounding tissue. While the role of the neutrophil is to promote host defense—very clearly established for bacteria and fungi, less clearly for viruses—neutrophils at the same time can promote tissue damage and destruction, representing the two sides of their classically defined “double-edged sword” (see reference 309 for a general review of the role of neutrophils in innate immunity). Eosinophils are also recruited to the lungs in response to primary hRSV bronchiolitis (133, 169), and both neutrophils and eosinophils are recruited to lung tissue in the PVM model of acute pneumovirus infection in vivo (88). Virus-specific cytotoxic T lymphocytes have been detected within 10 days of infection in human subjects (54, 181), and roles for CD4+ and CD8+ T lymphocytes in virus clearance have been inferred from rodent studies (81, 146, 147).

Proinflammatory cytokines have been detected in respiratory secretions and in bronchoalveolar lavage samples from individuals infected with hRSV (1, 169, 182, 263). Three of the major chemokines detected—IL-8, RANTES, and macrophage inflammatory protein 1α (MIP-1α)—have each been implicated in neutrophil and eosinophil chemotaxis and recruitment. MIP-1α has been of particular interest, since its levels have been shown to correlate with levels of eosinophil degranulation products (169) and with severity of disease (136).

Several recent studies have described a role for the pattern recognition receptors CD14 and/or toll-like receptor 4 (TL4) in innate host defense against hRSV in vivo (158, 170, 222). Among the findings, these studies documented persistence of hRSV virions in TLR-4-deficient mouse lungs, impaired NK cell trafficking and function, and reduced early NF-κB-dependent inflammatory responses. However, analogous experiments performed with TLR-4 deficient mice and the natural rodent paramyxovirus pathogen Sendai virus (mouse parainfluenza 1) yielded completely contradictory results (362). The use of natural host-pathogen pairs for the study of acute respiratory virus infection is discussed further in “Biochemical responses in mouse models” (below).

Modeling hRSV Infection in Cell Lines and in Experimental Animals

PVM.

The human pathogen, hRSV, and the mouse pathogen, PVM, can induce similar disorders in their natural hosts. As such, the mouse data that are available are compared and contrasted to what is currently understood about pneumovirus infection in a human host.

Target cells.

The primary targets of hRSV in vivo are respiratory epithelial cells, although infection of alveolar macrophages has been reported (278). This has been modeled in tissue culture, and the epithelial cell lines HEp-2 and A549 and primary human epithelial cultures have been used to study the cellular and molecular responses to hRSV infection. Other cells are susceptible to hRSV infection in culture, including the fibroblast cell line MRC-5, macrophage and macrophage-like cell lines (301, 313), and, for uptake if not outright infection, eosinophils (211). Mouse PVM has similar target affinity and infects respiratory epithelial cells in vivo and the mouse epithelial cell line LA4 (265).

Cellular receptors.

The receptor for PVM has not been characterized. While no one has yet identified a single specific protein, lipid, or carbohydrate entity that serves as a unique, virus-specific receptor for the hRSV pathogen, the ongoing story is an interesting one, with multiple virus and cell determinants coming into play. Among the earliest findings, Krusat and Streckert (218) demonstrated that the glycosaminoglycan (GAG) heparin, when included in an in vitro culture system, could inhibit the infection of target cells with hRSV. This finding has withstood the test of time, and several groups have followed through with significant elucidations and refinements. Taking the whole-virus approach, Hallak et al. (163, 164) reported that only N-sulfated moieties of GAGs (and not C6-O or C2-O sulfation) are important mediators of RSV infection and later documented the specific role of iduronic acid-substituted forms. This is consistent with the current overall thinking on GAGs as heterogeneous structures that promote specificity to a greater degree than initially appreciated. The reader is referred to several excellent reviews of GAG biology for a larger sense of the role of these carbohydrates in pathogen invasion (174, 238, 270, 330).

From the perspective of the virus, initial studies focused on the RSV G protein. Feldman et al. (115, 116) confirmed the interaction of the RSV G protein with heparin, identified a linear heparin binding domain within the G protein, and documented an interaction of heparin with the RSV F protein as well. However, Teng et al. (347) later demonstrated that deletion of the G-protein binding domain did not alter sensitivity to heparin, and this was followed by a study by Techaarpornkul et al. (341) demonstrating that recombinant RSV with deleted G and SH proteins not only was able to infect epithelial cells in culture but also was able to do so in a partially GAG-independent fashion. Teng et al. (347) also demonstrated that the membrane-bound form of the G protein was not absolutely required for replication in vitro and that the secreted form, without cytoplasmic and transmembrane domains, served adequately in its stead; a subsequent study (345) demonstrated that the conserved “cystine noose” region separating the two extracellular glycosylated domains of the hRSV G protein were likewise dispensible.

In summary, the issue of cellular receptors for hRSV remains one with many unanswered questions. GAGs clearly play a significant role in the infectivity of wild-type virus, and the recent identification of protein binding domains with high affinity for specific GAGs may be of some help in this endeavor (174). One of the most important questions that remains unanswered is that of species specificity; on that topic, a very recent and intriguing observation by Schlender et al. (316) documents the role of the F2 subunit domain in modulating the species specificity observed for human versus bovine RSV infection. Also of note is the report by Tripp et al. (353) on chemokine mimicry by the hRSV G protein, leading some to conclude that the chemokine receptor for fractalkine, CX3CR1, might serve as a coreceptor for hRSV infection. However, while epithelial cells synthesize and secrete fractalkine, there is no evidence for CX3CR1 expression in these major target cells for hRSV infection. Finally, the most recent report by Malhotra et al. (246) suggests a role for annexin II, which itself is a heparin binding protein and as such may interact with the glycosylated virus proteins and glycosylated target cell proteins in unexplored and unexpected ways.

Cellular and biochemical responses. (i) Syncytium formation.

The classic multicellular cytopathic syncytia formed in epithelial cell culture in response to infection with hRSV have been described in significant detail (195). While several lines of evidence suggest a role for the hRSV F protein in inducing syncytium formation, the molecular details of this morphologic response remain largely unknown. Recent evidence has demonstrated increased expression of the cytoskeleton filament protein cytokeratin-17 and its localization at sites of syncytium formation (91) as well as involvement of the small GTPase RhoA (281, 282).

(ii) Biochemical responses in cell culture.

The biochemical responses of epithelial cell cultures infected with hRSV have been reviewed recently (78, 92, 131); this will permit us to cover the highlights and focus here on the most recent studies. Among the most prominent biochemical responses of epithelial cells is the synthesis and secretion of the potent neutrophil chemoattractant IL-8. The transcriptional control of IL-8 relies on the activation of the master transcriptional activator, NF-κB (119, 135, 253, 255), with the involvement of NF-IL-6 (185, 254), ERK-2 (53), and unique hRSV-responding enhancer elements in the IL-8 gene promoter (48). hRSV infection of epithelial cells in vitro induces the production of other chemokines, including RANTES, monocyte chemotactic protein 1 (MCP-1), and MIP-1α, the last chemokine being of particular note because it is directly related to the inflammatory responses to both hRSV and PVM observed in vivo (90, 136, 169). Other prominent biochemical responses to hRSV infection in vitro include the production of inducible nitric oxide synthase and nitric oxide (155, 201, 356) beta interferon, IL-1α, IL-6, and IL-11 and increased expression of cell surface markers intercellular cell adhesion molecule 1 (ICAM-1; CD54), CD18, vascular cell adhesion molecule (VCAM), major histocompatibility complex classes I and II, CD14, and CD15 (reviewed in reference 92). See also the section on gene microarray approaches in “Gene microarray expression studies” below.

(iii) Biochemical responses in mouse models.

A number of groups have studied the pathogenesis of hRSV inoculation in various strains of inbred mice (primarily BALB/c) and cotton rats. All describe the moderate influx of inflammatory cells, with reports including lymphocytes and eosinophils as components of the respiratory infiltrates (120, 145, 147, 319, 320). Blanco et al. (25) documented the increased expression of transcripts encoding several cytokines, chemokines, alpha and gamma interferons, and the interferon response gene, IP-10, in response to inoculation of cotton rats with 105 PFU of hRSV-A Long strain. These results were similar to those reported by Haeberle et al. (157), who evaluated the responses of inoculated BALB/c mice, although the latter group also reported the inducible expression of MIP-1α. However, others have shown that while MIP-1α is produced in this model, the levels detected do not correlate with the extent of the inflammatory response (89), as has been observed in both human disease (136, 169) and the PVM mouse model (90).

This latter point leads us to an important caveat. It is important to understand that, just as there are two distinct disease entities studied in humans (discussed in “Inflammatory responses to infection” above), there are two distinct rodent models involving hRSV inoculation. The immunopotentiation model, in which mice are inoculated with live, attenuated, or fixed hRSV or hRSV components and then challenged with hRSV, is a model of acquired immunity and has been developed in conjunction with studies related to vaccine development. This application of hRSV needs to be distinguished from primary infection models, in which one is attempting to evaluate innate responses and innate immunity to infection. In many studies of the pathogenesis of primary infection in these nonnatural rat and mouse hosts, it is difficult to determine whether the responses observed are to the very limited virus replication observed or simply to large quantities of inoculated foreign antigens that happen to be viral in origin. The responses to infection and the responses to antigen bolus are not interchangeable, since they reflect different capacities and different reactivities of the innate inflammatory response. The remarkable evolutionary divergence of proteins involved in innate host defense (269) suggests that experiments relating to disease pathogenesis and innate host response need to be performed using appropriate species-pathogen pairs. Studies using natural host-pathogen pairs may be limited and limiting in other ways, but they ultimately provide better representations of the pathogenesis of pneumovirus infection in vivo.

PVM has recently been developed as an alternative model for the study of the pathogenesis of severe pneumovirus disease (27, 77, 88, 90, 93). Intransal inoculation of fewer than 30 PFU of virus results in clear-cut replication in mouse lung tissue, yielding titers as high as 108 PFU/g. Virus replication is accompanied by a profound inflammatory response, mucus production, and airway obstruction, leading to significant morbidity and mortality. As noted above, the inflammatory response is promoted by local production of the CC chemokine MIP-1α and its signaling through the receptor, CCR1, a pathway under exploration for combined antiviral-immunomodulatory therapy, as discussed in “Combination therapy” (below).

(iv) Gene microarray expression studies.

The gene microarray is a more global approach to the study of transcriptional responses to defined physiologic or pathophysiologic stimuli. As of this writing, three published studies have used this approach to evaluate the transcriptional responses of epithelial cells to hRSV in vitro and a fourth study has focused on the responses of whole mouse lung to infection with PVM in vivo. The first of these systematic studies, reported by Zhang et al. (390), presents an evaluation of the responses of the human epithelial cell line A549 and primary human SAE cells to infection with the A2 strain of RSV. The A549 responses included the anticipated increases in the levels of transcripts encoding the chemokines IL-8, RANTES, MCP-1, and MIP-1α, and have extended the list to include, among others, I-309 and fractalkine. Most interesting was the fact that the pattern observed in the primary SAE cells was not precisely identical, demonstrating the existence of distinct responses in what are often thought to be completely interchangeable model systems. The gene array study of the transcriptional responses in whole-mouse lung mRNA to infection with the pathogenic PVM strain J3666 likewise demonstrates increased expression of a specific array of proinflammatory mediators, including transcripts encoding murine MCP-1, RANTES, MCP-3, beta interferon, and 10 characterized interferon response and interferon-related genes (93). The second publication focused on the in vitro response (349) has provided us with a more general look at the responses directly related to the activation of the proinflammatory transcriptional master switch, NF-κB. Using a clever, dominant-negative mutant approach, the authors compared the responses of hRSV-A2-infected HeLa cells designed to express the IκBα-dominant negative mutant under doxycycline control and established that in addition to what we already know are NF-κB regulated transcripts (the IκB inhibitors and IL-8), several interferon response factors (IRF-1 and IRF-7B) and the transcription factor STAT-1 are also under NF-κB control. The third publication from this group (391) focuses on the antiviral agent ribavirin and its interference with hRSV- A2-mediated transcriptional activation of the A549 cell line. Among the conclusions, the authors determined that ribavirin, while an effective antiviral agent, actually does not interfere in a very substantial way with the inflammatory cascade initiated by hRSV replication, a result that has been echoed by the findings of Bonville et al. (27) in their study of the cellular inflammatory responses to PVM in vivo. Another important conclusion from the gene array study was that ribavirin administration resulted in enhanced expression of several components of the interferon-signaling pathway via as yet undefined interactions with the interferon-stimulated response enhancer elements common among these up-regulated transcripts (391), adding to our understanding of the more subtle immunomodulatory properties of ribavirin.

Variants and Mutants with Altered Pathogenicity

Natural variation.

Naturally occurring variants of hRSV fall into two diagnostic subgroups, hRSV-A and hRSV-B, initially identified on the basis of differential reactivity with monoclonal antibodies and later confirmed on the basis of gene sequence, with particular divergence noted within the sequence of the virus G gene (129). While the evolutionary significance of this divergence remains largely unknown, Woelk and Holmes (379) have identified six sites within the sequence of the hRSV G protein that have been subjected to positive selection, all within the extracellular domain and all associated to some degree with specific anti-hRSV immunoglobulin epitopes and/or glycosylation, suggesting immune-driven natural selection. The question of the relative pathogenicity of hRSV-A and hRSV-B variants has been addressed in a recent study by Martinello et al. (251) in which they found no association of disease severity with either the hRSV-A or hRSV-B subgroup (137 and 84 isolates, respectively), but went on to identify a specific A subgroup clade (GA3) with this characteristic. In contrast, a study of 28 isolates by Brandenburg et al. (33) found no relationship between hRSV-A lineages and disease severity. Further study is necessary to distinguish between these sets of results.

Tissue culture attenuation and random mutagenesis.

Prior to the development of reverse genetics, studies of the contributions of virus sequence to disease severity were dependent on the identification, isolation, and sequence analysis of naturally occurring virus variants, typically the result of the relaxation of functional constraints provided by replication in tissue culture. A good example of this comes from the PVM literature, specifically the study by Randhawa et al. (295) in which G-protein sequences of the mouse-passaged PVM strain J3666 and the tissue culture-passaged, attenuated PVM strain 15 were compared; the authors demonstrated different translational start sites leading to the absence of a putative amino-terminal cytoplasmic domain in the latter. The pace of discovery was enhanced by the addition of random mutagenesis methods, which led to the identification of important mutations in the P (phosphoprotein) (242) and L (polymerase) (351) proteins of hRSV.

Antibody escape mutants.

Escape mutants are essentially a technological form of immune-drive natural selection (see reference 261 for an excellent review and details of earlier studies). Escape mutants have been created from hRSV grown in tissue culture in the presence of neutralizing monoclonal antibodies directed against the virus G or F proteins (241, 252, 253, 368). This work has generated detailed antigenic maps of these proteins, has provided a virtual “snapshot” of the way in which hRSV virions respond to immunoglobulins in vivo, and has provided significant guidance on the development of passive immunoprophylaxis for hRSV (80).

Recombinant viruses.

Collins et al. (67) were the first to report the successful production of infectious hRSV from cloned cDNA in tissue culture. Cotransfection of HEp-2 cells with a plasmid containing the 15,223-nucleotide antigenome together with expression plasmids encoding the N, P, L, and M2 proteins permitted the recovery of infectious virions and led the way to the important next step, the creation of point, replacement, addition, and deletion mutations in the virus backbone. The use of this technology, known as reverse genetics, in studies of pneumovirus biology in general (68, 249) and its application to the vaccine effort (60, 69, 268) have recently been reviewed. With respect to the vaccine studies, among the more intriguing applications of this technology has been the inclusion of immunomodulatory cytokines IL-2 (40) and granulocyte-macrophage colony-stimulating factor (41) within the virus backbone in an attempt to alter the host response as part of the overall vaccination strategy. Deletion and substitution mutational analyses using this or alternative genome-based methods have led to a greater understanding of the role of various virus-encoded proteins (46, 188, 242, 339, 341), particularly that of F and G proteins in hRSV infectivity and species specificity (130, 316, 347, 392), described in detail in “Cellular receptors” (above).

Virus Evasion of Innate Immunity

RNA viruses use a large number and variety of mechanisms to subvert innate antiviral host defenses; many of those mechanisms involve evasion of interferon and interferon-regulated responses. Comparatively little is known about such strategies among the pneumoviruses (see references 143 and 385 for reviews discussing strategies used by other viruses of the family Paramyxoviridae). However, Atreya and Kulkarni (7) demonstrated that hRSV replication was not diminished in response to treatment with either alpha or beta interferon in several standard tissue culture model systems. Given that there is ample evidence for induced production of beta interferon by hRSV-infected epithelial cells (7, 134, 186, 356), the question of resistance remains acute. The most clear-cut evidence for resistance strategies to date comes from Conzelmann and colleagues (315), who demonstrated that the NS1 and NS2 proteins of bRSV and hRSV function coordinately and in a species-restricted fashion to antagonize the antiviral activities of alpha and beta interferons. Also, as noted above, bRSV NS-1 and NS-2 proteins inhibit the production of beta interferon by reducing the activation of interferon-regulatory factor 3 (28) and bRSV NS-2 specifically is involved in interferon production in bovine nasal fibroblasts and bronchoalveolar macrophages (358). Teng, Whitehead, and colleagues (346, 378) have demonstrated the attenuation of hRSV NS-1 and M2-2 or NS-1 and SII deletion mutants in the chimpanzee model of infection, although without specific reference to interferon-related phenomena. Interestingly, hRSV is sensitive to inhibition by gamma interferon (11, 20, 39, 99, 276, 365), and potential hRSV-mediated modulation pathways have beenidentified (6, 17).

Chemokine mimicry is another means by which viruses are known to subvert innate antiviral host defense. The potential role of the hRSV G protein in chemokine mimicry is an intriguing lead to be explored further (353). In another recent development, Zimmer et al. (394) describe virokinin, a post-translationally modified cleavage product of bRSV F protein that contains a classical motif identifying it as a member of the conserved tachykinin family of peptide hormones. The role of this virus-encoded protein in binding to tachykinin receptors in the respiratory tract and potentially altering the course of bRSV infection remains to be clarified.

Regarding virus evasion in vivo, the responses are complex and often difficult to evaluate or pinpoint with any assurance at the molecular level. One interesting interpretation of viral evasion in vivo comes from the work of Sakurai et al., (311), who isolated and evaluated naturally occurring antibodies reactive with the immunodominant F glycoprotein and found a subset of nonneutralizing antibodies with a high affinity for the immature, viral lysate forms of F, together with neutralizing subset with high affinity for mature F on infected cells and on virions. The authors consider the intriguing possibility that by evolving and maintaining two developmental forms of the F protein with distinct antigenicity, the virus can evade and subvert humoral immunity more effectively.

CLINICAL STRATEGIES

Passive Immunoprophylaxis for hRSV Disease

There are two formulations of anti-RSV immunoglobulins in use for prophylaxis against hRSV infection in high-risk patient groups. The earliest studies with experimental animals suggested that immunoglobulin preparations containing high-titer antibodies to hRSV (hRSV-Ig) protected against development of severe disease (150, 291, 323). While passive prophylaxis could attenuate the severity of hRSV disease, it did not prevent infection, and an increased number of unexplained deaths occurred in hRSV-Ig recipients with congenital heart disease, leading to the recommendation that RSV-Ig not be used in patients with this underlying condition (324); considerable controversy ensued regarding specific indications due primarily to the high cost of administering this agent (294). Other problems associated with the use of hRSV-Ig included the potential for transmission of blood-borne pathogens, the need to administer a large volume of product (15 ml/kg of body weight), and the associated fever and desaturation that can accompany the administration of any blood product.

In response to the need for a better prophylactic agent, a humanized monoclonal antibody against the hRSV F protein was developed for clinical use (reviewed in references 151 and 325). A randomized trial of humanized anti-F monoclonal antibody, palivizumab, was performed at 139 centers in North America and the United Kingdom, with intramuscular administration of this agent to high-risk infants once per month during hRSV season. This approach was shown to be highly effective in reducing hRSV-related hospitalizations (180a). Palivizumab was licensed by the Food and Drug Administration in June 1998. Emerging data demonstrating the safety of this monoclonal antibody product in patients with congenital heart disease will very probably lead to a Food and Drug Administration-approved indication for use in this group of high-risk individuals.

While hRSV-Ig and monoclonal anti-F antibody already play an important role in the prevention of serious hRSV infection, neither appears to be effective in the treatment of this disease. In a study of previously healthy infants randomized to receive RSV-Ig or placebo, no significant differences were observed in the duration of hospitalization, intensive care unit stay, mechanical ventilation, supplemental oxygen use, or adverse events between the two groups (306). One explanation for the lack of clinical efficacy during acute hRSV infection is that by the time respiratory symptoms are present, the virus is sequestered from host humoral immunity (294). Furthermore, gene microarray (391) and mouse model (27, 29) studies suggest that the inflammatory responses to pneumovirus infection are initiated very early on and that antiviral therapy alone is not effective in controlling the negative sequelae. Despite these observations, there is growing evidence that using palivizumab in transplant patients with documented hRSV upper respiratory tract infection may prevent the spread of infection to the lower lung when used in combination with ribavirin (85). This strategy is currently under investigation by the Collaborative Antiviral Study Group and may be of particular importance in this very high-risk patient population.

Active Immunoprophylaxis for hRSV Disease

The morbidity and mortality associated with hRSV bronchiolitis, coupled with its frequency and worldwide distribution, make it a prime target for the development of a vaccine that can be used early in life. There have been several forces at work that have hampered progress on a vaccine against hRSV. Most important is the possibility that vaccination will result in adverse reactions to naturally occurring hRSV disease, as observed with the formalin-inactivated vaccine used in the 1960s (55, 124, 202, 210). Furthermore, newborns and very young infants may not mount a protective immune response because of relative immunologic immaturity or suppression of their immune response due to circulating maternally derived anti-hRSV antibodies (68, 79, 108, 267). Natural hRSV infection itself elicits only partial immunity, and reinfection in later life is commonplace. Several strategies have been implemented recently in an attempt to generate safe and effective subunit, inactivated, and live-attenuated hRSV vaccines (60, 109). Currently, two promising candidate vaccines under study in clinical trials are an hRSV F subunit vaccine for immunization of patients who have already experienced primary RSV infection, such as the elderly and older hRSV-seropositive children with conditions predisposing them to severe hRSV disease (108, 279, 288, 354), and cold-passaged, temperature sensitive (cpts) attenuated hRSV strains (196, 206). cpts mutant virus strains are difficult, if not impossible, to “fine tune” by serial passage in tissue culture, emphasizing the importance of using newer recombinant (reverse genetics) technologies to advance the field. Recombinant hRSV vaccines under evaluation in clinical trials are largely based on the cpts 248/404 “backbone” virus, with molecular deletions and/or point mutations designed to enhance several desirable features. Another promising area involves hRSV vaccine strains that lack the NS1 and/or NS2 genes since deletion of these sequences augments the host cell interferon response pathways (31, 315).

Therapy for hRSV Disease

Antiviral agents.

While significant advances in our knowledge of hRSV biology, immunology, pathophysiology, and epidemiology have been made in the past 50 years, there continues to be a fair amount of controversy regarding the optimum management of infants and children with hRSV infection (369). Supportive therapy, including hospitalization, supplemental oxygen delivery, nasal and pulmonary toilet, and, in the most severe cases, intubation and mechanical ventilation, is the mainstay of clinical management.

(i) Ribavirin.

Lack of consensus regarding the use of ribavirin for moderate to severe forms of hRSV-induced bronchiolitis cannot be attributed to a paucity of clinical trials. Results from at least 14 trials of bronchodilators, 3 trials of adjunctive systemic glucocorticoid treatment, and 11 randomized trials of the antiviral synthetic nucleoside ribavirin have been published (16, 149, 159, 160, 229, 233, 259, 264, 294, 305, 327, 336, 369, 376). In six controlled, randomized studies, ribavirin treatment was found to reduce the severity of hRSV illness by decreasing viral shedding, improving oxygen saturation, and improving clinical scores (16, 159, 160, 207, 327, 336), but in 1996, a systematic overview of these studies highlighted potential flaws in study design and interpretation (296). The results of this meta-analysis were discouraging but were consistent with the clinical experience of pediatricians, specifically that ribavirin treatment alone did not lead to significant improvement in clinically important outcomes. The growing concern regarding the efficacy of ribavirin and the high cost of its use (118) led to a wording change in the American Academy of Pediatrics recommendation for its usage from “should be used” to “may be considered” for selected infants and young children at high risk for serious hRSV disease (70).

Severe hRSV infection during infancy is a known risk factor for the development of subsequent wheezing illnesses during the first decade of life. Studies of the outcome of bronchiolitis during infancy have included studies that evaluated the effects of ribavirin therapy during acute illness on the long-term sequelae of recurrent wheezing during childhood (101, 132, 208, 240, 256, 310), with promising results. In contrast, Krilov et al. (217) found no differences between ribavirin-treated and untreated infants in wheezing or other pulmonary function measures 6 to 8 years beyond their infection. Interestingly, new attention has also been raised regarding potential long-term benefits of using the leukotriene receptor antagonist montelukast during acute infection for the prevention of post-hRSV infection wheezing (24). A large multicenter clinical trial to test this hypothesis is under way.

(ii) New antiviral strategies: fusion inhibitors, RNases, and others.

New strategies for therapeutic intervention during moderate to severe hRSV disease are clearly needed. A variety of antiviral compounds have been tested for their ability to impair hRSV replication in vitro. Speculation that inhibiting cell-cell fusion (i.e., syncytium formation) might lessen the severity of clinical disease has led to attempts to develop hRSV-specific fusion inhibitors. Peptides derived from epitopes of the hRSV fusion or F protein blocked hRSV-induced syncytium formation at concentrations as low as 0.015 μM (226). Small-molecule hRSV-specific fusion inhibitors have neutralizing activity against A and B group hRSV isolates in vitro and in vivo via blockade of cell-cell fusion (179, 258, 272), while a benzodithiin compound has demonstrated ability to interfere with intracellular processing of the hRSV fusion protein, thereby resulting in decreased infectivity (333). Other agents reported to have anti-hRSV activity have included crude plant protein extracts (3), some forms of superoxide dismutase (384), an l-aspartate transcarbamoylase inhibitor (383), eosinophil-derived neurotoxin, a potent RNase (87), and lovastatin (144).

While nucleoside derivatives other than ribavirin have generated some interest by investigators in this field (359), enthusiasm for the development of antisense technology has been more robust. Specific cleavage of hRSV genome RNA has been detected at antisense oligonucleotide binding sites (184). Because of the pattern of RNA cleavage observed, it was suggested that endogenous RNase H participated in hRSV genomic degradation. Other RNases have been studied for their antiviral properties. For example, recruitment and activation of RNase L to an hRSV mRNA using 2-5A antisense oligonucleotide technology showed potent inhibition of hRSV replication in vitro (2) and, more recently, in vivo using an African green monkey model of hRSV infection (230). As of this writing, there are no publications describing antisense therapy in human clinical trials.

Combination therapy: antivirals with antibody, anti-inflammatory, and immunomodulatory strategies.

Clinically, the negative sequelae of pneumovirus infection involve active viral replication in conjunction with the host cell inflammatory response. Combination therapy with the antiviral ribavirin and systemically administered immunoglobulin has been tried with some success in the cotton rat model of hRSV infection (153), and one report of combined aerosolized ribavirin and systemic immunoglobulin treatment in bone marrow transplant recipients who developed hRSV disease showed a 22% mortality among patients treated before the onset of respiratory failure and 100% mortality in patients who went untreated or in whom treatment was initiated within 24 h of respiratory failure (377). Parenteral therapy with a monoclonal anti-F antibody in the cotton rat model of human hRSV infection resulted in clearance of hRSV within 24 h but had no effect on early pulmonary pathology, while glucocorticoid treatment reduced the inflammatory component of the pneumonitis but delayed virus clearance; when the two interventions were combined, an antiviral and anti-inflammatory effect was appreciated (292).

Increasing attention is being paid to the need for concurrent anti-inflammatory therapy for moderate to severe hRSV disease. To this end, glucocorticoids are generally considered to have broad-reaching anti-inflammatory properties and in fact have been the focus of several clinical hRSV trials. Unfortunately, these trials suggest that when used alone in the absence of antivirals, glucocorticoid treatment has only marginal, if any, benefit in the treatment of hRSV-infection and its associated inflammatory response (72, 83, 84, 212, 213, 231, 307, 331, 334, 338, 365). A meta-analysis of several these studies demonstrated that systemic glucocorticoids reduced the total length of hospital stay by only 0.43 day per patient, an effect reduced to only 0.29 day per patient when patients with previous episodes of wheezing were excluded from the analysis (137), a statistically significant difference with overall minor clinical impact. Interestingly, several recent studies showed that glucocorticoid treatment during hRSV bronchiolitis in vivo had no effect on hRSV titers measured in upper or lower respiratory secretions or on the release of the proinflammatory chemokine IL-8 (37; H. S. Jafri, C. M. Carubella, S. C. Buckingham, P. Sheeran, J. Hatfield, R. D. Hardy, J. P. DeVincenzo and O. Ramilo, Pediatr, Acad. Soc. Joint Meet, abstr. 1564, 2000) or on the release of IL-8 from hRSV-infected epithelial cells (28). The observation that glucocorticoids failed to blunt the virus-directed IL-8 response suggests the possibility that hRSV-infected epithelial cells may be resistant to the anti-inflammatory effects usually seen with steroid administration, an observation that was reproduced in mice infected with the natural pneumovirus pathogen PVM (94).

The approach involving a combined strategy that includes antiviral and agents directed at specific virus-induced proinflammatory events has been best characterized using the PVM mouse model (92, 94), described in detail in “Pathogenesis of disease” (above). The proinflammatory chemokine MIP-1α has been identified as an essential link between virus infection and granulocyte recruitment to infected lung tissue (90). However, once the virus infection is established, the continued production of MIP-1α and the cellular inflammatory response that ensues are not tightly linked to virus replication, and, while ribavirin has significant efficacy in reducing virus replication, it does nothing to ameliorate the virus-induced proinflammatory cascade (27). Dramatic clinical improvement was observed in response to systemic administration of ribavirin in combination with either neutralizing anti-MIP-1α antibody (27) or by pharmacologic blockade of the primary MIP-1α receptor CCR1 (29, 95). Translation of these laboratory findings may result in a clinically achievable therapeutic strategy for the management of severe hRSV infection in human infants.

CONCLUSIONS

Pneumovirus infection remains a significant problem for practitioners of both human and veterinary medicine. Advances in our understanding of pneumovirus biology have provided pathways toward new vaccine formulations. At the same time, advances in our understanding of the pathogenesis of pneumovirus disease in specific natural hosts have provided strategies for combined antiviral and anti-inflammatory/immunomodulatory therapies.

Acknowledgments

We are grateful to John Ellis, Department of Veterinary Microbiology, Western College of Veterinary Medicine at the University of Saskatchewan, Saskatoon, Canada, for his guidance in locating and interpreting relevant information from the veterinary literature.

REFERENCES

  • 1.Abu-Harb, M., F. Bell, A. Finn, W. H. Rao, L. Nixon, D. Shale, and M. L. Everard. 1999. IL-8 and neutrophil elastase levels in the respiratory tract of infants with RSV bronchiolitis. Eur. Respir. J. 14:139-143. [DOI] [PubMed] [Google Scholar]
  • 2.Adah, S. A., S. F. Bayly, H. Cramer, R. H. Silverman, and P. F. Torrence. 2001. Chemistry and biochemistry of 2′,5′-oligoadenylate-based strategy. Curr. Med. Chem. 8:1189-1212 [DOI] [PubMed] [Google Scholar]
  • 3.Ahmad, A., J. Davies, S. Randall, and G. R. Skinner. 1996. Antiviral properties of extract of Opuntia streptacantha. Antiviral Res. 30:75-85. [DOI] [PubMed] [Google Scholar]
  • 4.Ahmadian, G., P. Chambers and A. J. Easton. 1999. Detection and characterization of proteins encoded by the second ORF of the M2 gene of pneumoviruses. J. Gen. Virol. 80:2011-2016. [DOI] [PubMed] [Google Scholar]
  • 5.Alvarez, R., H. M. Lwamba, D. R. Kapczynski, M. K. Njenga, and B. S. Seal. 2003. Nucleotide and predicted amino acid sequence-based analysis of the avian metapneumovirus type C cell attachment glycoprotein gene: phylogenetic analysis and molecular epidemiology of the U.S. pneumoviruses. J. Clin. Microbiol. 41:1730-1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Aoyagi, M., N. Shimojo, K. Sekine, T. Nishimuta, and Y. Kohno. 2003. Respiratory syncytial virus infection suppresses IFN-gamma production of gamma-delta T cells. Clin. Exp. Immunol. 131:312-317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.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]
  • 8.Atreya, P. L., M. E. Peeples, and P. L. Collins. 1998. The NS1 protein of human respiratory syncytial virus is a potent inhibitor of minigenome transcription and RNA replication. J. Virol. 72:1452-1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bachi, T., and C. Howe. 1973. Morphogenesis and ultrastructure of respiratory syncytial virus. J. Virol. 12:1173-1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Baker, J. C., J. A. Ellis, and E. G. Clark. 1997. Bovine respiratory syncytial virus. Vet. Clin. North Am. Food Anim. Pract. 13:425-454. [DOI] [PubMed] [Google Scholar]
  • 11.Barends, M., A. Boelen, L. de Rond, J. Dormans, J. Kwakkel, X. van Oosten, J. H. Neijens, and T. G. Kimman. 2003. Respiratory syncytial virus enhances respiratory allergy in mice despite the inhibitory effect of virus-induced interferon gamma. J. Med. Virol. 69:156-162. [DOI] [PubMed] [Google Scholar]
  • 12.Barr, J., and A. J. Easton. 1995. Characterisation of the interaction between the nucleoprotein and phosphoprotein of pneumonia virus of mice. Virus Res. 39:221-235. [DOI] [PubMed] [Google Scholar]
  • 13.Barr, J., P. Chambers, C. R. Pringle, and A. J. Easton. 1991. Sequence of the major nucleocapsid protein gene of pneumonia virus of mice: sequence comparisons suggest structural homology between nucleocapsid proteins of pneumoviruses, paramyxoviruses, rhabdoviruses and filoviruses. J. Gen. Virol. 72:677-685. [DOI] [PubMed] [Google Scholar]
  • 14.Barr, J., P. Chambers, P. Harriott, C. R. Pringle, and A. J. Easton. 1994. Sequence of the phosphoprotein gene of pneumonia virus of mice: expression of multiple proteins from overlapping reading frames. J. Virol. 68:5330-5334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Barrett, T., D. K. Clarke, S. A. Evans, and B. K. Rima. 1987. The nucleotide sequence of the gene encoding the F protein of canine distemper virus: a comparison of the deduced amino acid sequence with other paramyxoviruses. Virus Res. 8:373-386. [DOI] [PubMed] [Google Scholar]
  • 16.Barry W., F. Cockburn, R. Cornall, J. F. Price, G. Sutherland, and A. Vardag. 1985. Ribavirin aerosol for acute bronchiolitis. Arch. Dis. Child. 61:593-597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bartz, H., O. Turkel, S. Hoffjan, T. Rothoeft, A. Gonschorek, and U. Schauer. 2003. Respiratory syncytial virus decreases the capacity of myeloid dendritic cells to induce interferon-gamma in naïve T cells. Immunology 109:49-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bastien, N., S. Normand, T. Taylor, D. Ward, T. C. Peret, G. Boivin, L. J. Anderson, and Y. Li. 2003. Sequence analysis of the N, P, M and F genes of Canadian human metapneumovirus strains. Virus Res. 93:51-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Begona Ruiz-Arguello, M., L. Gonzalez-Reyes, L. J. Calder, C. Aplomo, D. Martin, M. J. Saiz, B. Garcia-Barreno, J. J. Skehel, and J. A. Melero. 2002. Effect of proteolytic processing at two distinct sites on shape and aggregation of an anchorless fusion protein of human respiratory syncytial virus and fate of the intervening segment. Virology 298:317-326. [DOI] [PubMed] [Google Scholar]
  • 20.Behera, A. K., M. Kumar, R. F. Lockey, and S. S. Mohapatra. 2002. 2′-5′ oligoadenylate synthetase plays a critical role in interferon-gamma inhibition of respiratory syncytial virus infection of human epithelial cells. J. Biol. Chem. 277:25610-25608. [DOI] [PubMed] [Google Scholar]
  • 21.Bermingham, A., and P. L. Collins. 1999. The M2-2 protein of human respiratory syncytial virus is a regulatory factor involved in the balance between RNA replication and transcription. Proc. Natl. Acad. Sci. USA 96:11259-11264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Berthiaume, L., J. Joncas, and V. Pavilanis. 1974. Comparative structure, morphogenesis and biological characteristics of the respiratory syncytial (RS) virus and the pneumonia virus of mice (PVM). Arch. Gesamte Virusforsch. 45:39-51. [DOI] [PubMed] [Google Scholar]
  • 23.Bhella, D., A. Ralph, L. B. Murphy, and R. P. Yeo. 2002. Significant differences in nucleocapsid morphology within the Paramyxoviridae. J. Gen. Virol. 83:1831-1839. [DOI] [PubMed] [Google Scholar]
  • 24.Bisgaard, H., and Study Group on Montelukast and Respiratory Syncytial Virus. 2003. A randomized trial of montelukast in respiratory syncytial virus postbronchiolitis. Am. J. Respir. Crit. Care Med. 167:379-383. [DOI] [PubMed] [Google Scholar]
  • 25.Blanco, J. C. G., J. Y. Richardson, M. E. R. Darnell, A. Rowzee, L. Pletneva, D. D. Porter, and G. A. Prince. 2002. Cytokine and chemokine gene expression after primary and secondary respiratory syncytial virus infection in cotton rats. J. Infect. Dis. 185:1780-1785. [DOI] [PubMed] [Google Scholar]
  • 26.Boivin, G., Y. Abed, G. Pelletier, L. Ruel, D. Moisan, S. Cote, T. C. Peret, D. D. Erdman, and L. J. Anderson. 2002. Virological features and clinical manifestations associated with human metapneumovirus: a new paramyxovirus responsible for acute respiratory tract infections in all age groups. J. Infect. Dis. 186:1330-1334. [DOI] [PubMed] [Google Scholar]
  • 27.Bonville, C. A., A. J. Easton, H. F. Rosenberg, and J. B. Domachowske. 2003. Altered pathogenesis of severe pneumovirus infection in response to combined antiviral and specific immunomodulatory agents. J. Virol. 77:1237-1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bonville, C. A., P. A. Mehta, L. Krilov, H. F. Rosenberg, and J. B. Domachowske. 2001. Epithelial cells infected with respiratory syncytial virus are resistant to the anti-inflammatory effects of hydrocortisone. Cell. Immunol. 213:134-140. [DOI] [PubMed] [Google Scholar]
  • 29.Reference deleted
  • 30.Bossert, B., and K. K. Conzelmann. 2002. Respiratory syncytial virus (RSV) non- structural (NS) proteins as host range determinants: a chimeric bovine RSV with NS genes from human RSV is attenuated in interferon-competent bovine cells. J. Virol. 76:4287-4293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bossert, B., S. Marozin, and K. K. Conzelmann. 2003. Nonstructural proteins NS1 and NS2 of bovine respiratory syncytial virus block activation of interferon regulatory factor-3. J. Virol. 77:8661-8668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Brandenburg, A. H., H. J. Neijens, and A. D. Osterhaus. 2001. Pathogenesis of RSV lower respiratory tract infection: implications for vaccine development. Vaccine 19:2769-2782. [DOI] [PubMed] [Google Scholar]
  • 33.Brandenburg, A. H., R. van Beek, H. A. Moll, A. D. Osterhaus, and E. C. Claas. 2000. G protein variation in respiratory syncytial virus group A does not correlate with clinical severity. J. Clin. Microbiol. 38:3849-3852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Brogden, K. A., H. D. Lehmkuhl, and R. C. Cutlip. 1998. Pasteurella haemolytica complicated respiratory infections in sheep and goats. Vet. Res. 29:233-254. [PubMed] [Google Scholar]
  • 35.Buchholz, U. J., S. Finke, and K. K. Conzelmann. 1999. Generation of bovine respiratory syncytial virus (BRSV) from cDNA: BRSV NS2 is not essential for virus replication in tissue culture, and the human RSV leader region acts as a function BRSV genome promoter. J. Virol. 73:251-259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Buchholz, U. J., H. Granzow, K. Schuldt, S. S. Whitehead, B. R. Murphy, and P. L. Collins. 2000. Chimeric bovine respiratory syncytial virus with glycoprotein gene substitutions from human respiratory syncytial virus (HRSV): effects on host range and evaluation as a live-attenuated HRSV vaccine. J. Virol. 74:1187-1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Buckingham, S. C., H. S. Jafri, A. J. Bush, C. M. Carubelli, P. Sheeran, R. D. Hardy, M. G. Ottolini, O. Ramilio, and J. P. DeVincenzo. A randomized, double-blind, placebo-controlled trial of dexamethasone in severe respiratory syncytial virus (RSV) infection: effects on viral load and clinical outcomes. J. Infect. Dis. 185:1222-1228. [DOI] [PubMed]
  • 38.Bukreyev, A., S. S. Whitehead, B. R. Murphy and P. L. Collins. 1997. Recombinant respiratory syncytial virus from which the entire SH gene has been deleted grows efficiently in cell culture and exhibits site-specific attenuation in the respiratory tract of the mouse. J. Virol. 71:8973-8982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bukreyev, A., S. S. Whitehead, N. Bukreyeva, B. R. Murphy, and P. L. Collins. 1999. Interferon gamma expressed by a recombinant respiratory syncytial virus attenuates virus replication in mice without compromising immunogenicity. Proc. Natl. Acad. Sci. USA 96:2367-2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bukreyev, A., S. S. Whitehead, C. Prussin, B. R. Murphy, and P. L. Collins. 2000. Effect of coexpression of interleukin-2 by recombinant respiratory syncytial virus on virus replication, immunogenicity, and production of other cytokines. J. Virol. 74:7151-7157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bukreyev, A., I. M. Belyakov, J. A. Berzofsky, B. R. Murphy, and P. L. Collins. 2001. Granulocyte-macrophage colony-stimulating factor expressed by reocmbinant respiratory syncytial virus attenuates viral replication and increases the level of pulmonary antigen presenting cells. J. Virol. 75:12128-12140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bukreyev, A., B. R. Murphy, and P. L. Collins. 2000. Respiratory syncytial virus can tolerate an intergenic sequence of at least 160 nucleotides with little effect on transcription or replication in vitro and in vivo. J. Virol. 74:11017-11026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Calain, P., and L. Roux. 1993. The rule of six, a basic feature for efficient replication of Sendai virus defective interfering RNA. J. Virol. 67:4822-4830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cane, P. A., B. G. van den Hoogen, S. Chakrabarti, C. D. Fegan, and A. D. Osterhaus. 2003. Human metapneumovirus in a haematopoietic stem cell transplant recipient with fatal lower respiratory tract disease. Bone Marrow Transplant. 31:309-310. [DOI] [PubMed] [Google Scholar]
  • 45.Caravokyri, C., A. J. Zajac, and C. R. Pringle. 1992. Assignment of mutant tsN19 (complementation group E) of respiratory syncytial virus to the P protein gene. J. Gen. Virol. 73:865-873. [DOI] [PubMed] [Google Scholar]
  • 46.Cartee, T. L., and G. W. Wertz. 2001. Respiratory syncytial virus M2-1 protein requires phosphorylation for efficient function and binds viral RNA during infection. J. Virol. 75:12188-12197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cartee, T. L., A. G. Megaw, A. G. Oomens, and G. W. Wertz. 2003. Identification of a single amino acid change in the human respiratory syncytial virus L protein that affects transcriptional termination. J. Virol. 77:7352-7360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Casola, A., R. P. Garofalo, M. Jamaluddin, S. Vlahopoulos, and A. R. Brasier. 2000. Requirement of a novel upstream reponse element in respiratory syncytial virus-induced IL-8 gene expression. J. Immunol. 164:5944-5951. [DOI] [PubMed] [Google Scholar]
  • 49.Chambers, P., D. A. Matthews, C. R. Pringle, and A. J. Easton. 1990. The nucleotide sequences of intergenic regions between nine genes of pneumonia virus of mice establish the physical order of these genes in the viral genome. Virus Res. 18:263-270. [DOI] [PubMed] [Google Scholar]
  • 50.Chambers, P., C. R. Pringle, and A. J. Easton. 1991. Genes 1 and 2 of pneumonia virus of mice encode proteins which have little homology with the 1C and 1B proteins of human respiratory syncytial virus. J. Gen. Virol. 72:2545-2549. [DOI] [PubMed] [Google Scholar]
  • 51.Chambers, P., C. R. Pringle, and A. J. Easton. 1992. Sequence analysis of the gene encoding the fusion glycoprotein of pneumonia virus of mice suggests possible conserved secondary structure elements in paramyxovirus fusion glycoproteins. J. Gen. Virol. 73:1717-1724. [DOI] [PubMed] [Google Scholar]
  • 52.Champlin, R. E., and E. Whimbey. 2001. Community respiratory virus infections in bone marrow transplant recipients: the M. D. Anderson Cancer Center experience. Biol. Blood Marrow Transplant. 7(Suppl.):8S-10S. [DOI] [PubMed] [Google Scholar]
  • 53.Chen, W., M. M. Monick, A. B. Carter, and G. W. Hunninghake. 2000. Activation of ERK2 by respiratory syncytial virus in A549 cells is linked to the production of interleukin-8. Exp. Lung Res. 26:13-26. [DOI] [PubMed] [Google Scholar]
  • 54.Chiba, Y., Y. Higashidato, and K. Suga. 1989. Development of cell-mediated cytotoxic immunity to respiratory syncytial virus in human infants following naturally acquired infection. J. Med. Virol. 28:133-139. [DOI] [PubMed] [Google Scholar]
  • 55.Chin J., R. L. Maggoffin, L. A. Shearer, J. H. Schieble, and E. H. Lennette. 1969. Field evaluation of a respiratory syncytial virus vaccine and a trivalent parainfluenza virus vaccine in a pediatric population. Am. J. Epidemiol. 89:449-463. [DOI] [PubMed] [Google Scholar]
  • 56.Choi, E. H., H. J. Lee, T. Yoo, and S. J. Chanock. 2002. A common haplotype of interleukin-4 gene IL4 is associated with severe respiratory syncytial virus disease in Korea. J. Infect. Dis. 186:1207-1211. [DOI] [PubMed] [Google Scholar]
  • 57.Collins, P. L., and G. W. Wertz. 1985. Nucleotide sequences of the 1B and 1C nonstructural protein mRNAs of human respiratory syncytial virus. Virology 143:442-451. [DOI] [PubMed] [Google Scholar]
  • 58.Collins, P. L., and G. W. Wertz. 1985. The 1A protein gene of human respiratory syncytial virus: nucleotide sequence of the mRNA and a related polycistronic transcript. Virology 141:283-291. [DOI] [PubMed] [Google Scholar]
  • 59.Collins, P. L., and G. Mottet. 1993. Membrane orientation and oligomerization of the small hydrophobic protein of human respiratory syncytial virus. J. Gen. Virol. 74:1445-1450. [DOI] [PubMed] [Google Scholar]
  • 60.Collins, P. L., and B. R. Murphy. 2002. Respiratory syncytial virus: reverse genetics and vaccine strategies. Virology 296:204-211. [DOI] [PubMed] [Google Scholar]
  • 61.Collins, P. L., L. E. Dickens, A. Buckler-White, R. A. Olmsted, M. K. Spriggs, E. Camargo, and K. V. Coelingh. 1986. Nucleotide sequences for the gene junctions of human respiratory syncytial virus reveal distinctive features of intergenic structure and gene order. Proc. Natl. Acad. Sci. USA 83:4594-4598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Collins, P. L., M. G. Hill, E. Camargo, H. Grosfeld, R. M. Chanock, and B. R. Murphy. 1995. Production of infectious human respiratory syncytial virus from cloned cDNA confirms an essential role for the transcription elongation factor from the 5′ proximal open reading frame of the M2 mRNA in gene expression and provides a capability for vaccine development. Proc. Natl. Acad. Sci. USA 92:11563-11567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.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]
  • 64.Collins, P. L., Y. T. Huang, and G. W. Wertz. 1984. Nucleotide sequence of the gene encoding the fusion (F) glycoprotein of human respiratory syncytial virus. Proc. Natl. Acad. Sci. USA 81:7683-7687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Collins, P. L., R. A. Olmsted, M. K. Spriggs, P. R. Johnson, and A. J. Buckler- White. 1987. Gene overlap and site-specific attenuation of transcription of the viral polymerase L gene of human respiratory syncytial virus. Proc. Natl. Acad. Sci. USA 84:5134-5138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Collins, P. L., M. A. Mink, and D. S. Stec. 1991. Rescue of synthetic analogs of respiratory syncytial virus genomic RNA and effect of truncations and mutations on the expression of a foreign reporter gene. Proc. Natl. Acad. Sci. USA 88:9663-9667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Collins, P. L., M. G. Hill, E. Camargo, H. Grosfeld, R. M. Chanock, and B. R. Murphy. 1995. Reproduction of infectious human respiratory syncytial virus from cloned cDNA confirms an essential role for the transcription elongation factor from the 5′ proximal open reading frame of the M2 mRNA in gene expression and provides a capability for vaccine development. Proc. Natl. Acad. Sci. USA 92:11563-11567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Collins, P. L., R. M. Chanock, B. R. Murphy. 2001. Respiratory syncytial virus, p. 1443-1486. In D. M. Knipe and P. M. Howley (ed.) Fields virology, 4th ed., vol. 1. Lippincott-Raven Publishers, Philadelphia, Pa.
  • 69.Collins, P. L., S. S. Whitehead, A. Bukreyev, R. Fearns, M. N. Teng, K. Juhasz, R. M. Chanock, and B. R. Murphy. 2002. Rational design of live-attenuated recombinant vaccine virus for human respiratory syncytial virus by reverse genetics. Adv. Virus Res. 54:423-451. [DOI] [PubMed] [Google Scholar]
  • 70.Committee on Infectious Disease, American Academy of Pediatrics. 1996. Reassessment of the indications for ribavirin therapy in respiratory syncytial virus infections. Pediatrics 97:137-140. [PubMed] [Google Scholar]
  • 71.Compans, R. W., D. H. Harter, and P. W. Choppin. 1967. Studies of pneumonia virus of mice (PVM) in cell culture. II. Structure and morphogenesis of the virus particle. J. Exp. Med. 126:267-277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Connolly, J. H., C. M. B. Field, J. F. T. Glasgow, C. M. Slattery, and D. M. MacLynn. 1969. A double blind trial of prednisolone in epidemic bronchiolitis due to respiratory syncytial virus. Acta Pediatr. Scand. 58:116-120. [DOI] [PubMed] [Google Scholar]
  • 73.Cook, J. K. 2000. Avian pneumovirus infections of turkeys and chickens. Vet. J. 160:118-125. [DOI] [PubMed] [Google Scholar]
  • 74.Cook, J. K., and D. Cavanagh. 2002. Detection and differentiation of avian pneumoviruses (metapneumoviruses). Avian Pathol. 31:117-132. [DOI] [PubMed] [Google Scholar]
  • 75.Cook, J. K., M. M. Ellis, and M. B. Huggins. 1991. The pathogenesis of turkey rhinotracheitis virus in turkey poults inoculated with the virus alone or together with two strains of bacteria. Avian Pathol. 20:155-166. [DOI] [PubMed] [Google Scholar]
  • 76.Cook, J. K., S. Kinloch, and M. M. Ellis. 1993. In vitro and in vivo studies in chickens and turkeys on strains of turkey rhinotracheitis virus isolated from the two species. Avian Pathol. 22:157-170. [DOI] [PubMed] [Google Scholar]
  • 77.Cook, P. M., R. P. Eglin, and A. J. Easton. 1998. Pathogenesis of pneumovirus infections in mice: detection of pneumonia virus of mice and human respiratory syncytial virus mRNA in lungs of infected mice by in situ hybridization. J. Gen. Virol. 79:2411-2417. [DOI] [PubMed] [Google Scholar]
  • 78.Crowe, J. E., Jr. 1999. Host responses to respiratory syncytial virus infection and immunization. Curr. Top. Microbiol. Immunol. 236:191-214. [DOI] [PubMed] [Google Scholar]
  • 79.Crowe, J. E., Jr. 2001. Influence of maternal antibodies on neonatal immunization against respiratory viruses. Clin. Infect. Dis. 33:1720-1727. [DOI] [PubMed] [Google Scholar]
  • 80.Crowe, J. E., C. Y. Firestone, R. Crim, J. A. Beeler, K. L. Coelingh, C. F. Barbas, D. R. Burton, R. M. Chanock, and B. R. Murphy. 1998. Monoclonal antibody-resistant mutants selected with a respiratory syncytial virus neutralizing human antibody Fab fragment (Fab 19) define a unique epitope on the fusion (F) glycoprotein. Virology 20:373-375. [DOI] [PubMed] [Google Scholar]
  • 81.Culley, F. J., J. Pollott, and P. J. Openshaw. 2002. Age at first viral infection determines the pattern of T-cell-mediated disease during reinfection in adulthood. J. Exp. Med. 196:1381-1386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Curran, J., and D. Kolakofsky. 1999. Replication of paramyxoviruses. Adv. Virus Res. 54:403-422. [DOI] [PubMed] [Google Scholar]
  • 83.Dabbous, I. A., J. S. Tkachyk, and S. J. Stamm. 1966. A double blind study of the effects of corticosteroids in the treatment of bronchiolitis. Pediatrics 37:477-484. [PubMed] [Google Scholar]
  • 84.De Boeck, K., N. van der Aa, S. van Lierde, L. Corbeel, and R. Eeckels. 1997. Respiratory syncytial virus bronchiolitis: a double blind dexamethasone efficacy study. J. Pediatr. 131:919-921. [DOI] [PubMed] [Google Scholar]
  • 85.DeVincenzo, J. P., R. L. Hirsch, R. J. Fuentes, and F. H. Top, Jr. 2000. Respiratory syncytial virus immune globulin treatment of lower respiratory tract infection in pediatric patients undergoing bone marrow transplantation—a compassionate use experience. Bone Marrow Transplant. 25:161-165. [DOI] [PubMed] [Google Scholar]
  • 86.Domachowske, J. B., and H. F. Rosenberg. 1999. Respiratory syncytial virus infection: immune response, immunopathogenesis and treatment. Clin. Microbiol. Rev. 12:298-309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Domachowske, J. B., K. D. Dyer, C. A. Bonville, and H. F. Rosenberg. 1998. Recombinant human eosinophil-derived neurotoxin/ RNase 2 functions as an effective antiviral agent against respiratory syncytial virus. J. Infect. Dis. 177:1458-1464. [DOI] [PubMed] [Google Scholar]
  • 88.Domachowske, J. B., C. A. Bonville, K. D. Dyer, A. J. Easton, and H. F. Rosenberg. 2000. Pulmonary eosinophilia and production of MIP-1α are prominent responses to infection with pneumonia virus of mice. Cell. Immunol. 200:98-104. [DOI] [PubMed] [Google Scholar]
  • 89.Domachowske, J. B., C. A. Bonville, J. L. Gao, P. M. Murphy, A. J. Easton, and H. F. Rosenberg. 2000. MIP-1α is produced but it does not control pulmonary inflammation in response to respiratory syncytial virus in mice. Cell. Immunol. 206:1-6. [DOI] [PubMed] [Google Scholar]
  • 90.Domachowske, J. B., C. A. Bonville, J. L. Gao, P. M. Murphy, A. J. Easton, and H. F. Rosenberg. 2000. The chemokine macrophage-inflammatory protein-1α and its receptor CCR1 control pulmonary inflammation and antiviral host defense in paramyxovirus infection. J. Immunol. 165:2677-2682. [DOI] [PubMed] [Google Scholar]
  • 91.Domachowske, J. B., C. A. Bonville, and H. F. Rosenberg. 2000. Cytokeratin 17 is expressed in cells infected with respiratory syncytial virus via NF-κB activation and is associated with the formation of cytopathic syncytia. J. Infect. Dis. 182:1022-1028. [DOI] [PubMed] [Google Scholar]
  • 92.Domachowske, J. B., C. A. Bonville, and H. F. Rosenberg. 2001. Gene expression in epithelial cells in response to pneumovirus infection. Respir. Res. 2:225-233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Domachowske, J. B., C. A. Bonville, A. J. Easton, and H. F. Rosenberg. 2002. Differential expression of proinflammatory cytokine genes in vivo in response to pathogenic and nonpathogenic pneumovirus infections. J. Infect. Dis. 186:8-14. [DOI] [PubMed] [Google Scholar]
  • 94.Domachowske, J. B., C. A. Bonville, D. Ali-Ahmad, K. D. Dyer, A. J. Easton, and H. F. Rosenberg. 2001. Glucocorticoid administration accelerates mortality of pneumovirus-infected mice. J. Infect. Dis. 184:1518-1523. [DOI] [PubMed] [Google Scholar]
  • 95.Domachowske, J. B., C. A. Bonville, and H. F. Rosenberg. 2003. Successful therapeutic intervention for severe bronchiolitis using pharmacologic blockade of chemokine receptor-1 in combination with antiviral therapy. Pediatr. Res. A1917.
  • 96.Dorman, S. E., G. Uzel, J. Roesler, J. S. Bradley, J. Bastian, G. Billman, X. King, A. Filie, J. Schermerhorn, and S. M. Holland. 1999. Viral infections in interferon-gamma receptor deficiency. J. Pediatr. 135:640-643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Duncan, R. B., Jr., and L. N. Potgieter. 1993. Antigenic diversity of respiratory syncytial viruses and its implication for immunoprphylaxis in ruminants. Vet. Microbiol. 37:319-341. [DOI] [PubMed] [Google Scholar]
  • 98.Dupuy, L. C., S. Dobson, V. Bitko, and S. Barik. 1999. Casein kinase 2- mediated phosphorylation of respiratory syncytial virus phosphoprotein P is essential for the transcription elongation activity of the viral polymerase; phosphorylation by casein kinase 1 occurs mainly at Ser 215 and is without effect. J. Virol. 73:8384-8392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Durbin, J. E., T. R. Johnson, R. K. Durbin, S. E. Mertz, R. A. Morotti, R. S. Peebles, and B. S. Graham. 2002. The role of IFN in respiratory syncytial virus pathogenesis. J. Immunol. 168:2944-2952. [DOI] [PubMed] [Google Scholar]
  • 100.Easton, A. J., and P. Chambers. 1997. Nucleotide sequence of the genes encoding the matrix and small hydrophobic proteins of pneumonia virus of mice. Virus Res. 48:27-33. [DOI] [PubMed] [Google Scholar]
  • 101.Edell, D., V. Khoshoo, G. Ross, and K. Salter. 2002. Early ribavarin treatment of bronchiolitis: effect on long-term respiratory morbidity. Chest 122:935-939. [DOI] [PubMed] [Google Scholar]
  • 102.Ellis, J., K. West, C. Konoby, T. Leard, G. Gallo, J. Conlon, and N. Fitzgerald. 2001. Efficacy of an inactivated respiratory syncytial virus vaccine in calves. J. Am. Vet. Med. Assoc. 218:1973-1980. [DOI] [PubMed] [Google Scholar]
  • 103.Ellis, J. A. 2001. The immunology of the bovine respiratory disease complex. Vet. Clin. North Am. Food Anim. Pract. 17:535-550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Elvander, M., S. Vilcek, C. Baule, A. Uttenthal, A. Ballagi-Pordany, and X. Belak. 1998. Genetic and antigenic analysis of the G attachment protein of bovine respiratory syncytial virus strains. J. Gen. Virol. 79:2939-2946. [DOI] [PubMed] [Google Scholar]
  • 105.Esper, F., D. Boucher, C. Weibel, R. A. Martinello, and J. S. Kahn. 2003. Human metapneumovirus infection in the United States: clinical manifestations associated with a newly emerging respiratory infection in children. Pediatrics 111:1407-1410. [DOI] [PubMed] [Google Scholar]
  • 106.Evans, J. E., P. A. Cane, and C. R. Pringle. 1996. Expression and characterization of the NS1 and NS2 proteins of respiratory syncytial virus. Virus Res. 43:155-161. [DOI] [PubMed] [Google Scholar]
  • 107.Everard, M. L., A. Swarbrick, M. Wrightham, J. McIntyre, C. Dunkley, P. D. James, H. F. Sewell, and A. D. Milner. 1994. Analysis of cells obtained by bronchial lavage of infants with respiratory syncytial virus infection. Arch. Dis. Child. 71:428-432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Falsey, A. R., and E. E. Walsh. 1996. Safety and immunogenicity of a respiratory syncytial virus subunit vaccine (PFP-2) in ambulatory adults over age 60. Vaccine 14:1214-1218. [DOI] [PubMed] [Google Scholar]
  • 109.Falsey, A. R., and E. E. Walsh. 1997. Safety and immunogenicity of a respiratory syncytial virus subunit vaccine (PFP-2) in the institutionalized elderly. Vaccine 15:1130-1132. [DOI] [PubMed] [Google Scholar]
  • 110.Falsey, A. R., D. Erdman, L. J. Anderson, and E. E. Walsh. 2003. Human metapneumovirus infections in young and elderly adults. J. Infect. Dis. 187:785-790. [DOI] [PubMed] [Google Scholar]
  • 111.Fearns, R., and P. L. Collins. 1999. Model for polymerase access to the overlapped L gene of respiratory syncytial virus. J. Virol. 73:388-397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.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]
  • 113.Fearns, R., M. E. Peeples, and P. L. Collins. 1997. Increased expression of the N protein of respiratory syncytial virus stimulates minigenome replication but does not alter the balance between the synthesis of mRNA and antigenome. Virology 236:188-201. [DOI] [PubMed] [Google Scholar]
  • 114.Fearns, R., M. E. Peeples, and P. L. Collins. 2002. Mapping the transcription and replication promoters of respiratory syncytial virus. J. Virol. 76:1663-1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Feldman, S. A., R. M. Hendry, and J. A. Beeler. 1999. Identification of a linear heparin binding domain for human respiratory syncytial virus attachment protein G. J. Virol. 73:6610-6617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Feldman, S. A., A. Audet, and J. A. Beeler. 2000. The fusion glycoprotein of human respiratory syncytial virus facilitates virus attachment and infectivity via an interaction with cellular heparan sulfate. J. Virol. 74:6442-6447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.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]
  • 118.Feldstein, T., J. Swegarden, and G. Atwwod. 1995. Ribavirin therapy: implementation of hospital guidelines and effect on usage and cost of therapy. Pediatrics 96:14-17. [PubMed] [Google Scholar]
  • 119.Fielder, M. A., K. Wernke-Dollries, and J. M. Stark. 1996. Inhibition of viral replication reverses respiratory syncytial virus-induced NF-κB activation and interleukin-8 gene expression in A549 cells. J. Virol. 70:9079-9082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Fischer, J. E., J. E. Johnson, R. K. Kuli-Zade, T. R. Johnson, S. Aung, R. A. Parker, and B. S. Graham. 1997. Overexpression of interleukin-4 delays virus clearance in mice infected with respiratory syncytial virus. J. Virol. 71:8672-8677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Fogg, M. H., K. R. Parsons, L. H. Thomas, and G. Taylor. 2001. Identification of CD4+ T cell epitopes on the fusion (F) and attachment (G) proteins of bovine respiratory syncytial virus (BRSV) Vaccine 19:3226-3240. [DOI] [PubMed] [Google Scholar]
  • 122.Follett, E. A., C. R. Pringle, and T. H. Pennington. 1975. Virus development in enucleate cells: echovirus, poliovirus, pseudorabies virus, reovirus, respiratory syncytial virus and Semliki Forest virus. J. Gen. Virol. 26:183-196. [DOI] [PubMed] [Google Scholar]
  • 123.Freymouth, F., A. Vabret, L. Legrand, N. Eterrandossi, F. Lafay-Delaire, J. Brouard, and B. Guillois. 2003. Presence of the new human metapneumovirus in French children with bronchitis. Pediatr. Infect. Dis. J. 22:92-94. [DOI] [PubMed] [Google Scholar]
  • 124.Fulginetti, V. A., J. J. Eller, O. F. Seiber, J. W. Joyner, M. Minamitani, and G. Meiklejohn. 1969. Respiratory virus immunization. I. A field trial of two inactivated respiratory virus vaccines: an aqueous trivalent parainfluenza virus vaccine and an alum-precipitated respiratory syncytial virus vaccine. Am. J. Epidemiol. 89:435-448. [DOI] [PubMed] [Google Scholar]
  • 125.Gaddum, R. M., R. S. Cook, J. M. Furze, S. A. Ellis, and G. Taylor. 2003. Recognition of bovine respiratory syncytial virus proteins by bovine CD8+ T lymphocytes. Immunology 108:220-229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Garcia, J., B. Garcia-Barreno, A. Vivo, and J. A. Melero. 1993. Cytoplasmic inclusions of respiratory syncytial virus-infected cells: formation of inclusion bodies in transfected cells that co-express the nucleoprotein, the phosphoprotein, and the 22K protein. Virology 195:243-247. [DOI] [PubMed] [Google Scholar]
  • 127.Garcia-Barreno, B., T. Delgado, and J. A. Melero. 1994. Oligo(A) sequences of human respiratory syncytial virus G protein gene: assessment of their genetic stability in frameshift mutants. J. Virol. 68:5460-5468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Garcia-Barreno, B., T. Delgado, and J. A. Melero. 1996. Identification of protein regions involved in the interaction of human respiratory syncytial virus phosphoprotein and nucleoprotein: significance for nucleocapsid assembly and formation of cytoplasmic inclusions. J. Virol. 70:801-808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Garcia-Barreno, B., C. Palomo, C. Penas, T. Delgado, P. Perez-Brena, and, J. A. Melero. 1989. Marked differences in the antigenic structure of human respiratory syncytial virus F and G glycoproteins. J. Virol. 63:925-932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Garcia-Beato, R., and J. A. Melero. 2000. The C-terminal third of human respiratory syncytial virus attachment (G) protein is partially resistant to protease digestion and is glycosylated in a cell-type-specific manner. J. Gen. Virol. 81:919-927. [DOI] [PubMed] [Google Scholar]
  • 131.Garofalo, R. P., and H. Haeberle. 2000. Epithelial regulation of innate immunity to respiratory syncytial virus. Am. J. Respir. Cell Mol. Biol. 23:581-585. [DOI] [PubMed] [Google Scholar]
  • 132.Garofalo, R. P., R. C. Welliver, and P. Ogra. 1989. Modulation of leukotriene (LT) release with ribavirin during infection with respiratory syncytial virus. Pediatr. Res. 25:163a. [Google Scholar]
  • 133.Garofalo, R. P., J. L. L. Kimpen, R. C. Welliver, and P. L. Ogra. 1992. Eosinophil degranulation in the respiratory tract during naturally acquired respiratory syncytial virus infection. J. Pediatr. 120:28-32. [DOI] [PubMed] [Google Scholar]
  • 134.Garofalo, R. P., 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-regulated class I MHC expression through the induction of interferon-beta and IL-1α. J. Immunol. 157:2506-2513. [PubMed] [Google Scholar]
  • 135.Garofalo, R. P., M. Sabry, M. Jamaluddin, R, K. Yu, A. Casola, P. L. Ogra, and A. R. Brasier. 1996. Transcriptional activation of the interleukin-8 gene by respiratory syncytial virus infection in alveolar epithelial cells: nuclear translocation of the RelA transcription factor as a mechanism producing airway mucosal inflammation. J. Virol. 70:8773-8781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Garofalo, R. P., J. Patti, K. A. Hintz, V. Hill, P. L. Ogra, and R. C. Welliver. 2001. Macrophage inflammatory protein-1alpha (not T helper type 2 cytokines) is associated with severe forms of respiratory syncytial virus bronchiolitis. J. Infect. Dis. 184:393-399. [DOI] [PubMed] [Google Scholar]
  • 137.Garrison, M. M., D. A. Christakis, E. Harvey, P. Cummings, and R. L. Davis. 2000. Systemic corticosteroids in infant bronchiolitis: a meta-analysis. Pediatrics 105:E44. [DOI] [PubMed] [Google Scholar]
  • 138.Gershwin, L. J., E. S. Schelegle, R. A. Gunther, M. L. Anderson, A. R. Woolums, D. R. Larochelle, G. A. Boyle, K. E. Friebertshauser, and R. S. Singer. 1998. A bovine model of vaccine enhanced respiratory syncytial virus pathophysiology. Vaccine 16:1225-1236. [DOI] [PubMed] [Google Scholar]
  • 139.Gershwin, L. J., R. A. Gunther, M. L. Anderson, A. R. Woolums, K. McArthur-Vaughan, K. E. Randel, G. A. Boyle, K. E. Friebertshauser, and P. S. McInturff. 2000. Bovine-respiratory syncytial virus-specific IgE is associated with interleukin-2 and -4 and interferon-gamma expression in pulmonary lymph of experimentally infected calves. Am. J. Vet. Res. 61:291-298. [DOI] [PubMed] [Google Scholar]
  • 140.Ghildyal, R., J. Mills, M. Murray, N. Vardaxis, and J. Meanger. 2002. Respiratory syncytial virus matrix protein associates with nucleocapsids in infected cells. J. Gen. Virol. 83:753-757. [DOI] [PubMed] [Google Scholar]
  • 141.Goldmann, D. A. 2000. Transmission of viral respiratory infections in the home Pediatr. Infect. Dis. J. 19:S97-S102. [DOI] [PubMed] [Google Scholar]
  • 142.Gonzalez-Reyes, L., M. B. Ruiz-Arguello, B. Garcia-Barreno, L. Calder, J. A. Lopez, J. P. Albar, J. J. Skehel, D. C. Wiley, and J. A. Melero. 2001. Cleavage of the human respiratory syncytial virus fusion protein at two distinct sites is required for activation of membrane fusion. Proc. Natl. Acad. Sci. USA 98:9859-9864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Gotoh, B., T. Komatsu, K. Takeuchi, and J. Yokoo. 2001. Paramyxovirus accessory proteins as interferon antagonists. Microbiol. Immunol. 45:787-800. [DOI] [PubMed] [Google Scholar]
  • 144.Gower, T. L., and B. S. Graham. 2001. Antiviral activity of lovastatin against respiratory syncytial virus in vivo and in vitro Antimicrob. Agents Chemother. 45:1231-1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Graham, B. S., M. D. Perkins, P. F. Wright, and D. T. Karzon. 1988. Primary respiratory syncytial infection in mice. J. Med. Virol. 26:153-162. [DOI] [PubMed] [Google Scholar]
  • 146.Graham, B. S., L. A. Bunton, and P. F. Wright. 1991. Reinfection of mice with respiratory syncytial virus. J. Med. Virol. 34:7-13. [DOI] [PubMed] [Google Scholar]
  • 147.Graham, B. S., L. A. Bunton, P. F. Wright, and D. T. Karzon. 1991. Role of T-lymphocyte 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]
  • 148.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]
  • 149.Groothuis, J. R., K. A. Woodin, and R. Katz. 1990. Early ribavirin treatment of respiratory syncytial viral infection in high-risk children. J Pediatr. 117:792-798. [DOI] [PubMed] [Google Scholar]
  • 150.Groothuis, J. R., E. A. F. Simoes, and M. J. Levin. 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]
  • 151.Groothuis, J. R., and H. Nishida. 2002. Prevention of respiratory syncytial virus infections in high-risk infants by monoclonal antibody (palivizumab). Pediatr. Int. 44:235-241. [DOI] [PubMed] [Google Scholar]
  • 152.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]
  • 153.Gruber, W. C., S. Z. Wilson, B. J. Throop, and P. R. Wyde. 1987. Immunoglobulin administration and ribavirin therapy: efficacy in respiratory syncytial virus infection in the cotton rat. Pediatr. Res. 21:270-274. [DOI] [PubMed] [Google Scholar]
  • 154.Gulati, B. R., D. P. Patnayak, A. M. Sheikh, P. E. Poss, and S. M. Goyal. 2001. Protective efficacy of high-passage avian pneumovirus (APV/MN/turkey/1-a/97) in turkeys. Avian Dis. 45:593-597. [PubMed] [Google Scholar]
  • 155.Hacking, D., K. Rockett, J. Hull, and D. Kwiatkowski. 2002. Synergistic action of cytokines and purified respiratory syncytial virus in nitric oxide induction. J. Leukoc. Biol. 71:729-730. [PubMed] [Google Scholar]
  • 156.Hacking, D., and J. Hull. 2002. Respiratory syncytial virus—viral biology and the host response. J. Infect. 45:18-24. [DOI] [PubMed] [Google Scholar]
  • 157.Haeberle, H. A., W. A. Kuziel, J. H. Dieterich, A. Casola, Z. Gatalica, and R. P. Garofalo. 2001. Inducible expression of inflammatory chemokines in respiratory syncytial virus-infected mice: role of MIP-1α in lung pathology. J. Virol. 75:878-890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Haeberle, H. A., R. Takizawa, A. Casola, A. R. Brasier, H. J. Dieterich, N. Van Rooijen, Z. Gatalica, and R. P. Garofalo. 2002. Respiratory syncytial virus-induced activation of nuclear factor-kappa B in the lung involves alveolar macrophages and toll-like receptor 4-dependent pathways. J. Infect. Dis. 186:1199-1206. [DOI] [PubMed] [Google Scholar]
  • 159.Hall, C. B., J. T. McBride, and E. E. Walsh. 1983. Aerosolized ribavirin treatment of infants with respiratory syncytial virus infection. N. Engl. J. Med. 308:1443-1447. [DOI] [PubMed] [Google Scholar]
  • 160.Hall, C. B., J. T. McBride, C. L. Gala, S. W. Hildreth, and K. C. Schnabel. 1985. Ribavirin treatment of respiratory syncytial viral infection in infants with underlying cardiopulmonary disease. JAMA 254:3047-3051. [PubMed] [Google Scholar]
  • 161.Hall, C. B. 2000. Nosocomial respiratory syncytial virus infections: the “Cold War” has not ended. Clin. Infect. Dis. 31:590-596. [DOI] [PubMed] [Google Scholar]
  • 162.Hall, C. B. 2001. Respiratory syncytial virus and parainfluenza virus. N. Engl. J. Med. 344:1917-1928. [DOI] [PubMed] [Google Scholar]
  • 163.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]
  • 164.Hallak, L. K., D. Spillmann, P. L. Collins, and M. E. Peeples. 2000. Glycosaminoglycan sulfation requirements for respiratory syncytial virus infection. J. Virol. 74:10508-10513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.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]
  • 166.Hardy, R. W., and G. W. Wertz. 2000. The Cys3-His1 motif of respiratory syncytial virus M2-1 protein is essential for virus function. J. Virol. 74:5880-5885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Hardy, R. W., S. B. Harmon, and G. W. Wertz. 1999. Diverse gene junctions of respiratory syncytial virus modulate the efficiency of transcription termination and respond differently to M2-mediated antitermination. J. Virol. 73:170-176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Harmon, S. B., A. G. Megaw, and G. W. Wertz. 2001. RNA sequences involved in transcriptional termination of respiratory syncytial virus. J. Virol. 75:36-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Harrison, A. M., C. A. Bonville, H. F. Rosenberg, and J. B. Domachowske. 1999. Respiratory syncytial virus-induced chemokine expression in the lower airways: eosinophil recruitment and degranulation. Am. J. Respir. Dis. Crit. Care Med. 159:1918-1924. [DOI] [PubMed] [Google Scholar]
  • 170.Haynes, L. M., D. D. Moore, E. A. Kurt-Jones, R. W. Finberg, L. J. Anderson, and R. A. Tripp. 2001. Involvement of toll-like receptor 4 in innate immunity to respiratory syncytial virus. J. Virol. 75:10730-10737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Heegaard, P. M., D. L. Godson, M. J. Toussaint, K. Toornehooj, L. E. Larsen, B. Viuff, and L. Roonsholt. 2000. The acute phase response of haptoglobin and serum amyloid A (SAA) in cattle undergoing experimental infection with bovine respiratory syncytial virus. Vet. Immunol. Immunopathol. 23:151-159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Heminway, B. R., Y. Yu, Y. Tanaka, K. G., Perrine, E. Gustafson, J. M. Bernstein, and M. S. Galinski. 1994. Analysis of respiratory syncytial virus F, G, and SH proteins in cell fusion. Virology 200:801-805. [DOI] [PubMed] [Google Scholar]
  • 173.Hengst, U., and P. Kiefer. 2000. Domains of human respiratory syncytial virus P protein essential for homodimerisation and for binding to N and NS1 protein. Virus Genes 20:221-225. [DOI] [PubMed] [Google Scholar]
  • 174.Hileman, R. E., J. R. Fromm, J. M. Weiler, and R. J. Linhardt. 1998. Glycosaminoglycan-protein interactions: definition of consensus sites in glycosaminoglycan binding proteins. Bioessays 20:156-157. [DOI] [PubMed] [Google Scholar]
  • 175.Hoebee, B., E. Rietveld, L. Bont, M. Oosten, H. M. Hodemaekers, N. J. Nagelkert, H. J. Heijens, J. L. Kimpen, and T. G. Kimman. 2003. Association of severe respiratory syncytial virus bronchiolitis with interleukin-4 and interleukin-4 receptor alpha polymorphisms. J. Infect. Dis. 187:2-11. [DOI] [PubMed] [Google Scholar]
  • 176.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]
  • 177.Hull, J., A. Thomson, and D. Kwiatkowski. 2000. Association of respiratory syncytial virus bronchiolitis with the interleukin 8 gene region in UK families. Thorax 55:1023-1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Hull, J., H. Ackerman, K. Isles, S. Sen, M. Pinder, A. Thomson, and D. Kwiatkowski. 2001. Unusual haplotypic structure of IL8, a susceptibility locus for a common respiratory virus. Am. J. Hum. Genet. 69:413-419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Huntley, C. C., W. J. Weiss, A. Gazumyan, A. Buklan, B. Field, W. Hu, T. R. Jones, T. Murphy, A. A. Nikitenko, B. O'Hara, G. Prince, S. Quartuccio, Y. E. Raifeld, P. Wyde, and J. F. O'Connell. 2002. RFI-641, a potent respiratory syncytial virus inhibitor. Antimicr. Agents Chemother. 46:841-847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Hussell, T., and P. J. Openshaw. 1998. The effect of IL-12 treatment on vaccine-enhanced illness during infection with respiratory syncytial virus. Dev. Biol. Stand. 92:179-185. [PubMed] [Google Scholar]
  • 180a.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]
  • 181.Isaacs, D., C. R. M. Bangham, and A. J. McMichael. 1987. Cell-mediated cytotoxic response to respiratory syncytial virus in infants with bronchiolitis. Lancet ii:769-771. [DOI] [PubMed] [Google Scholar]
  • 182.Jafri, H. S. 2002. Role of chemokines in respiratory syncytial virus disease. Pediatr. Infect. Dis. J. 21:454-456. [DOI] [PubMed] [Google Scholar]
  • 183.Reference deleted.
  • 184.Jairath, S., P. B. Vargas, H. A. Hamlin, A. K. Field, and R. E. Kilkuskie. 1997. Inhibition of respiratory syncytial virus replication by antisense oligodeoxyribonucleotides. Antiviral Res. 33:201-213. [DOI] [PubMed] [Google Scholar]
  • 185.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]
  • 186.Jamaluddin, M., S. Wang, R. P. Garofalo, T. Elliott, A. Casola, S. Baron, A. R. Brasier. 2001. IFN-beta mediates coordinate expression of antigen-processing genes in RSV-infected pulmonary epithelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 280:L248-L257. [DOI] [PubMed] [Google Scholar]
  • 187.Jartti, T., B. van den Hoogen, R. P. Garofalo, A. D. Osterhaus, and O. Ruuskanen. 2002. Metapneumovirus and acute wheezing in children. Lancet 360:1393-1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Jin, H., H. Zhou, X. Cheng, R. Tang, M. Munoz, and N. Nguyen. 2000. Recombinant respiratory syncytial viruses with deletions in the NS1, NS2, SH and M2-2 genes are attenuated in vitro and in vivo. Virology 273:210-218. [DOI] [PubMed] [Google Scholar]
  • 189.Jirjis, F. F., S. L. Noll, D. A. Halvorson, K. V. Nagaraja, and D. P. Shaw. 2002. Pathogenesis of avian pneumovirus infection in turkeys. Vet. Pathol. 39:300-310. [DOI] [PubMed] [Google Scholar]
  • 190.Johnson, T. R., and B. S. Graham. 1999. Secreted respiratory syncytial virus G glycoprotein induces interleukin-5 (IL-5), IL-13, and eosinophilia by an IL-4-independent mechanism. J. Virol. 73:8485-8495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Johnson, T. R., J. E. Johnson, S. R. Roberts, G. W. Wertz, R. A. Parker, and B. S. Graham. 1998. Priming with secreted glycoprotein G of respiratory syncytial virus (RSV) augments interleukin-5 production and tissue eosinophilia after RSV challenge. J. Virol. 72:2871-2880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Joncas, J., L. Berthiaume, and V. Pavilanis. 1969. The structure of the respiratory syncytial virus. Virology 38:493-496. [DOI] [PubMed] [Google Scholar]
  • 193.Jones, R. C., C. Baxter-Jones, C. E. Savage, D. F. Kelly, and G. P. Wilding. 1987. Experimental infection of chickens with ciliostatic agent isolated from turkeys with rhinotracheitis. Vet. Rec. 120:301-302. [DOI] [PubMed] [Google Scholar]
  • 194.Jones, R. C., R. A. Williams, C. Baxter-Jones, C. E. Savage, and G. P. Wilding. 1988. Experimental infection of laying turkeys with rhinotracheitis virus: distribution of virus in the tissues and serological response. Avian Pathol. 17:841-850. [DOI] [PubMed] [Google Scholar]
  • 195.Jordon, W. S., Jr. 1962. Growth characteristics of respiratory syncytial virus. J. Immunol. 88:581-590. [PubMed] [Google Scholar]
  • 196.Juhasz, K., S. S. Whitehead, C. A. Boulanger, C. Y. Firestone, P. L. Collins, and B. R. Murphy. 1999. The two amino acid substitutions in the L protein of cpts530/1009, a live-attenuated respiratory syncytial virus candidate vaccine, are independent temperature-sensitive and attenuation mutations. Vaccine 17:1416-1424. [DOI] [PubMed] [Google Scholar]
  • 197.Kahn, J. S. 2003. Human metapneumovirus: a newly emerging respiratory pathogen. Curr. Opin. Infect. Dis. 16:255-258. [DOI] [PubMed] [Google Scholar]
  • 198.Kahn, J. S. 2003. Human metapneumovirus, a newly emerging respiratory virus. Pediatr. Infect. Dis. J. 22:921-924. [DOI] [PubMed] [Google Scholar]
  • 199.Kahn, J. S., M. J. Schnell, L. Buonocore, and J. K. Rose. 1999. Recombinant vesicular stomatitis virus expression respiratory syncytial virus (RSV) glycoproteins: RSV fusion protein can mediate infection and cell fusion. Virology 254:81-91. [DOI] [PubMed] [Google Scholar]
  • 200.Kamps, M. P., S. S. Taylor, and B. M. Sefton. 1984. Direct evidence that oncogenic tyrosine kinases and cyclic AMP-dependent protein kinase have homologous ATP binding sites. Nature 310:589-592. [DOI] [PubMed] [Google Scholar]
  • 201.Kao, Y. J., P. A. Piedra, G. L. Larsen, and G. N. Colasurdo. 2001. Induction and regulation of nitric oxide syntase in airway epithelial cells by respiratory syncytial virus. Am. J. Respir. Crit. Care Med. 163:532-539. [DOI] [PubMed] [Google Scholar]
  • 202.Kapikian, A. Z., R. H. Mitchell, R. M. Chanock, R. A. Shvedoff, and C. E. Stewart. 1969. An epidemiologic study of altered reactivity to respiratory syncytial (RS) virus infection in children previously vaccinated with an inactivated RS virus vaccine. Am. J. Epidemiol. 89:405-421. [DOI] [PubMed] [Google Scholar]
  • 203.Karaca, K., J. M. Sharma, M. A. Tomai, and R. L. Miller. 1996. In vivo and in vitro interferon induction in chickens by S-28828, an imidazquinolinamine immunoenhancer. J. Interferon Cytokine Res. 16:327-332. [DOI] [PubMed] [Google Scholar]
  • 204.Karger, A., U. Schmidt, and U. J. Buchholz. 2001. Recombinant bovine respiratory syncytial virus with deletions of the G or SH genes: G and F proteins bind heparin. J. Gen. Virol. 82:631-640. [DOI] [PubMed] [Google Scholar]
  • 205.Karron, R. A., D. A. Buonagurio, A. F. Georgiu, S. S. Whitehead, J. E. Adamus, M. L. Clements-Mann, D. O. Harris, V. B. Randolph, S. A. Udem, B. R. Murphy, and M. S. Sidhu. 1997. Respiratory syncytial virus (RSV) SH and G proteins are not essential for viral replication in vitro: clinical evaluation and molecular characterization of a cold-passaged, attenuated RSV subgroup B mutant. Proc. Natl. Acad. Sci. USA 94:13961-13966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Karron, R. A., P. F. Wright, J. E. Crowe, Jr., M. L. Clements-Mann, J. Thompson, M. Makhene, R. Casey, and B. R. Murphy. 1997. Evaluation of two live, cold passaged, temperature-sensitive respiratory syncytial virus (RSV) vaccines in chimpanzees, and in human adults, infants and children. J. Infect. Dis. 176:1428-1436. [DOI] [PubMed] [Google Scholar]
  • 207.Khattar, S. K., A. S. Yunus, and S. K. Samal. 2001. Mapping the domains on the phosphoprotein of bovine respiratory syncytial virus required for N-P and P-L interactions using a minigenome system. J. Gen. Virol. 82:775-779. [DOI] [PubMed] [Google Scholar]
  • 208.Khoshoo, V., G. Ross, and D. Edell. 2002. Effect of interventions during acute respiratory syncytial virus bronchiolitis on subsequent long term respiratory morbidity. Pediatr. Infect. Dis. J. 21:468-472. [DOI] [PubMed] [Google Scholar]
  • 209.Kim, C. K., C. Y. Chung, S. J. Choi, D. K. Kim, Y. Park, and Y. Y. Koh. 2000. Bronchoalveloar lavage cellular composition in acute asthma and acute bronchiolitis. J. Pediatr. 137:517-522. [DOI] [PubMed] [Google Scholar]
  • 210.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]
  • 211.Kimpen, J. L., R. Garofalo, R. C. Welliver, K. Fujihara, and P. L. Ogra. 1996. An ultrastructural study of the interaction of human eosinophils with respiratory syncytial virus. Pediatr. Allergy Immunol. 7:48-53. [DOI] [PubMed] [Google Scholar]
  • 212.Klassen, T. P., T. Sutcliffe, L. K. Watters, G. A. Wells, U. D. Allen, and M. Li. 1977. Dexamethasone in salbutamol-treated inpatients with acute bronchiolitis: a randomized controlled trial. J. Pediatr. 130:191-196. [DOI] [PubMed] [Google Scholar]
  • 213.Klassen, T. P., W. R. Craig, and D. Moher. 1998. Nebulized budesonide and oral dexamethasone for treatment of croup. JAMA 279:1629-1632. [DOI] [PubMed] [Google Scholar]
  • 214.Knott, I., V., K. Weynants, W. H. Walravens, J. H. van der Poel, J. A. Kramps, and J. J. Letesson. 1998. Immune response of calves experimentally infected with non-cell-culture-passaged bovine respiratory syncytial virus. Arch. Virol. 143:1119-1128. [DOI] [PubMed] [Google Scholar]
  • 215.Krempl, C., B. R. Murphy, and P. L. Collins. 2002. Recombinant respiratory syncytial virus with the G and F genes shifted to the promoter-proximal positions. J. Virol. 76:11931-11942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Krilov, L. R. 2001. Respiratory syncytial virus: update on infection, treatment and prevention. Curr. Infect. Dis. Rep. 3:242-246. [DOI] [PubMed] [Google Scholar]
  • 217.Krilov, L. R., F. S. Mandel, and S. R. Barone. 1997. Follow-up of children with respiratory syncytial virus bronchiolitis in 1986 and 1987: potential effect of ribavirin on long term pulmonary function. Pediatr. Infect. Dis. J. 16:273-276. [DOI] [PubMed] [Google Scholar]
  • 218.Krusat, T., and H. J. Streckert. 1997. Heparin-dependent attachment of respiratory syncytial virus (RSV) to host cells. Arch. Virol. 142:1247-1254. [DOI] [PubMed] [Google Scholar]
  • 219.Kuo, L., R. Fearns, and P. L. Collins. 1996. The structurally diverse intergenic regions of respiratory syncytial virus do not modulate sequential transcription by a dicistronic minigenome. J. Virol. 70:6143-6150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Kuo, L., H. Grosfeld, J. Cristina, M. G. Hill, and P. L. Collins. 1996. Effect of mutations in the gene-start and gene-end sequence motifs on transcription of monocistronic and dicistronic minigenomes of respiratory syncytial virus. J. Virol. 70:6892-6901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Kuo, L., R. Fearns, and P. L. Collins. 1997. Analysis of the gene start and gene end signals of human respiratory syncytial virus: quasi-templated initiation at position 1 of the encoded mRNA. J. Virol. 71:4944-4953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.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 meidate responses to respiratory syncytial virus. Nat. Immunol. 1:398-401. [DOI] [PubMed] [Google Scholar]
  • 223.Lahti, M., J. Lofgren, R. Marttila, M. Renko, T. Klaavuniemi, R. Haataja, M. Ramet, and M. Hallman. 2002. Surfactant protein D gene polymorphism associated with severe respiratory syncytial virus infection. Pediatr. Res. 51:696-699. [DOI] [PubMed] [Google Scholar]
  • 224.Lamb, R. A., and D. Kolakofsky. 1996. Paramyxoviridae: the viruses and their replication, p. 1177-1204. In B. N. Fields, D. M. Knipe, and P. M. Howley (ed.), Virology, 3rd ed. Lippincott-Raven, Philadelphia, Pa.
  • 225.Lambden, P. R. 1985. Nucleotide sequence of the respiratory syncytial virus phosphoprotein gene. J. Gen. Virol. 66:1607-1612. [DOI] [PubMed] [Google Scholar]
  • 226.Lambert, D. M., S. Barney, A. L. Lambert, K. Guthrie, R. Medinas, T. Bucy, J. Erickson, G. Merutka, and S. R. Petteway, Jr. 1996. Peptides from conserved regions of paramyxovirus fusion (F) proteins are potent inhibitors of viral fusion. Proc. Natl. Acad. Sci. USA 93:2186-2191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Larsen, L. E. 2000. Bovine respiratory syncytial virus (BRSV): a review. Acta Vet. Scand. 41:1-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Larsen, L. E., K. Tjornehoj, and B. Viuff. 2000. Extensive sequence divergence among bovine respiratory syncytial viruses isolated during recurrent outbreaks in closed herds. J. Clin. Microbiol. 38:4222-4227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Law, B. J., E. E. Wang, N. MacDonald, J. McDonald, S. Dobson, F. Boucher, J. Langley, J. Robinson, L. Mitchell, and D. Stephens. 1997. Does ribavirin impact on the hospital course of children with respiratory syncytial virus infection? An analysis using the pediatric investigators collaborative network on infections in Canada (PICNIC) RSV database. Pediatrics 99:E7. [DOI] [PubMed] [Google Scholar]
  • 230.Leaman, D. W., F. J. Longano, J. R. Okicki, K. F. Soike, P. F. Torrence, R. H. Silverman, and H. Cramer. 2002. Targeted therapy of respiratory syncytial virus in African green monkeys by intranasal administered 2-5A antisense. Virology 292:70-77. [DOI] [PubMed] [Google Scholar]
  • 231.Leer, J. A., J. L. Green, and E. M. Heimlich. 1969. Corticosteroid treatment in bronchiolitis. Am. J. Dis. Child. 117:495-503. [PubMed] [Google Scholar]
  • 232.Levine, S., R. Klaiber-Franco, and P. R. Paradiso. 1987. Demonstration that glycoprotein G is the attachment protein of respiratory syncytial virus. J. Gen. Virol. 68:2521-2524. [DOI] [PubMed] [Google Scholar]
  • 233.Lewinsohn, D. M., R. A. Bowden, D. Mattson, and S. W. Crawford. 1996. Phase 1 study of intravenous ribavirin treatment of respiratory syncytial virus pneumonia after marrow transplantation. Antimicrob. Agents Chemother. 40:2555-2557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Li, J., R. Ling, J. S. Randhawa, K. Shaw, P. J. Davis, K. Juhasz, C. R. Pringle, A. J. Easton, and D. Cavanagh. 1996. Sequence of the nucleocapsid protein gene of subgroup A and B avian pneumoviruses. Virus Res. 41:185-191. [DOI] [PubMed] [Google Scholar]
  • 235.Ling, R., and C. R. Pringle. 1989. Polypeptides of pneumonia virus of mice. II. Characterization of the glycoproteins. J. Gen. Virol. 70:1441-1452. [DOI] [PubMed] [Google Scholar]
  • 236.Ling, R., P. J. Davis, Q. Yu, C. M. Wood, C. R. Pringle, D. Cavanagh, and A. J. Easton. 1995. Sequence and in vitro expression of the phosphoprotein gene of avian pneumovirus. Virus Res. 36:247-257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Ling, R., A. J. Easton, and C. R. Pringle. 1992. Sequence analysis of the 22K, SH and G genes of turkey rhinotracheitis virus and J. Gen. Virol. 73:1709-1715. [DOI] [PubMed] [Google Scholar]
  • 238.Liu, J., and S. C. Thorp. 2002. Cell surface heparan sulfate and its roles in assisting viral infections. Med. Res. Rev. 22:1-25. [DOI] [PubMed] [Google Scholar]
  • 239.Lofgren, J., M. Ramet, M. Renko, R. Marttila, and M. Hallman. 2002. Association between surfactant protein A gene locus and severe respiratory syncytial virus infection in infants. J. Infect. Dis. 185:283-289. [DOI] [PubMed] [Google Scholar]
  • 240.Long, C. E., K. Z. Voter, W. Barker, and C. Hall. 1997. Long term follow-up of children hospitalized with respiratory syncytial virus lower respiratory tract infection and randomly treated with ribavirin or placebo. Pediatr. Infect. Dis. J. 16:1023-1028. [DOI] [PubMed] [Google Scholar]
  • 241.Lopez, J. A., R. Bustos, C. Orvell, M. Berois, J. Arbiza, B. Garcia-Barreno, and J. A. Melero. 1998. Antigenic structure of human respiratory syncytial virus fusion glycoprotein. J. Virol. 72:6922-6928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Lu, B., C. H. Ma, R. Brazas, and H. Jin. 2002. The major phosphorylation sites of the respiratory syncytial virus phosphoprotein are dispensable for virus replication in vitro. J. Virol. 76:10776-10784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Lu, B., R. Brazas, C. H. Ma, T. Kirstoff, X. Cheng, and H. Jin. 2002. Identification of temperature-sensitive mutations in the phosphoprotein of respiratory syncytial virus that are likely involved in its interaction with the nucleoprotein. J. Virol. 76:2871-2880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Lwamba, H. C., R. S. Bennett, D. C. Lauer, D. A. Halvorson, and M. K. Njenga. 2002. Characterization of avian metapnuemoviruses isolated in the USA. Anim. Health Res. Rev. 3:107-117. [DOI] [PubMed] [Google Scholar]
  • 245.Mackay, I. M., K. C. Jacob, D. Woolhouse, K. Waller, M. W. Syrmis, D. M. Whiley, D. J. Siebert, M. Nissen, and T. P. Sloots. 2003. Molecular assays for detection of human metapneumovirus. J. Clin. Microbiol. 41:100-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Malhotra, R., M. Ward, H. Bright, R. Priest, M. R. Foster, M. Hurle, E. Blair, and M. Bird. 2003. Isolation and characterization of potential respiratory syncytial virus receptor(s) on epithelial cells. Microbes Infect. 5:123-133. [DOI] [PubMed] [Google Scholar]
  • 247.Malik Peiris, J. S., W.-H. Tang, K.-H. Chan, P.-L., Khong, Y. Guan, Y.-L. Lau, and S. S. Chiu. 2003. Children with respiratory disease associated with metapneumovirus in Hong Kong. Emerg. Infect. Dis. 9:628-633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Mallipeddi, S. K., B. Lupiani, and S. K. Samal. 1996. Mapping the domains on the phosphoprotein of bovine respiratory syncytial virus required for N-P interaction using a two-hybrid system. J. Gen. Virol. 77:1019-1023. [DOI] [PubMed] [Google Scholar]
  • 249.Marriott, A. C., and A. J. Easton. 1999. Reverse genetics of the Paramyxoviridae. Adv. Virus Res. 53:321-340. [PubMed] [Google Scholar]
  • 250.Marriott, A. C., J. M. Smith, and A. J. Easton. 2001. Fidelity of leader and trailer sequence usage by the respiratory syncytial virus and avian pneumovirus replication complexes. J. Virol. 75:6265-6272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Martinello, R. A., M. D. Chen, C. Weibel, and J. S. Kahn. 2002. Correlation between respiratory syncytial virus genotype and severity of illness. J. Infect. Dis. 186:839-842. [DOI] [PubMed] [Google Scholar]
  • 252.Martinez, I., J. Dopazo and J. A. Melero. 1997. Antigenic structure of the human respiratory syncytial virus G glycoprotein and relevance of hypermutation events for the generation of antigenic variants. J. Gen. Virol. 78:2419-2429. [DOI] [PubMed] [Google Scholar]
  • 253.Martinez, I., and J. A. Melero. 2002. A model for the generation of multiple A to G transitions in the human respiratory syncytial virus genome: predicted RNA secondary structures as substrates for adenosine deaminases that act on RNA. J. Gen. Virol. 83:1445-1455. [DOI] [PubMed] [Google Scholar]
  • 254.Mastronarde, J. G., B. He, M. M. Monick, N. Mukaida, K. Matsushima, and G. W. Hunninghake. 1996. Induction of interleukin (IL)-8 gene expression by respiratory syncytial virus involved activation of nuclear factor (NF)-κB and NF-IL-6. J. Infect. Dis. 174:262-267. [DOI] [PubMed] [Google Scholar]
  • 255.Mastronarde, J. G., M. M. Monick, N. Mukaida, K. Matsushima, and G. W. Hunninghake. 1998. Activator protein-1 is the preferred transcription factor for cooperative interaction with nuclear factor-κB in respiratory syncytial virus-induced interleukin-8 gene expression in airway epithelium. J. Infect. Dis. 177:1275-1281. [DOI] [PubMed] [Google Scholar]
  • 256.McConnochie, K. M., J. D. Mark, J. T. McBride, W. J. Hall, and J. G. Brooks. 1985. Normal pulmonary function measurements and airway reactivity in childhood after mild bronchiolitis. Pediatrics 107:54-58. [DOI] [PubMed] [Google Scholar]
  • 257.McInnes, E., R. A. Collins, and G. Taylor. 1998. Cytokine expression in pulmonary and peripheral blood mononuclear cells from calves infected with bovine respiratory syncytial virus. Res. Vet. Sci. 64:163-166. [DOI] [PubMed] [Google Scholar]
  • 258.McKimm-Breschkin, J. 2000. VP-14637 Viropharma. Curr. Opin. Investig. Drugs 1:425-427. [PubMed] [Google Scholar]
  • 259.Meert, K. L., A. P. Sarnaik, M. J. Gelmini, and M. W. Lieh-Lai. 1994. Aerosolized ribavirin in mechanically ventilated children with respiratory syncytial virus lower respiratory tract disease: a prospective, double blind, randomized trial. Crit. Care Med. 22:566-572. [DOI] [PubMed] [Google Scholar]
  • 260.Meissner, H. C. 2003. Selected populations at increased risk from respiratory syncytial virus infection. Pediatr. Infect. Dis. J. 22:S40-S44. [DOI] [PubMed] [Google Scholar]
  • 261.Melero, J. A., B. Garcia-Barreno, L. Martinez, C. R. Pringle, and P. A. Cane. 1997. Antigenic structure, evolution and immunobiology of human respiratory syncytial virus attachment (G) protein. J. Gen. Virol. 78:2411-2418. [DOI] [PubMed] [Google Scholar]
  • 262.Mink, M. A., D. S. Stec, and P. L. Collins. 1991. Nucleotide sequences of the 3′ leader and 5′ trailer regions of human respiratory syncytial virus genomic RNA. Virology 185:615-624. [DOI] [PubMed] [Google Scholar]
  • 263.Mobbs, K. J., R. L. Smyth, U. O'Hea, D. Ashby, P. Ritson, and C. A. Hart. 2002. Cytokines in severe respiratory syncytial virus bronchiolitis. Pediatr. Pulmonol. 33:449-452. [DOI] [PubMed] [Google Scholar]
  • 264.Moler, F. W., C. M. Steinhart, S. E. Ohmit, G. L. Stidham, and the Pediatric Critical Care Study Group. 1996. Effectiveness of ribavirin in otherwise well infants with respiratory syncytial virus-associated respiratory failure. J. Pediatr. 128:422-428. [DOI] [PubMed] [Google Scholar]
  • 265.Moreau, J. M., K. D. Dyer, C. A. Bonville, T. Nitto, N. L. Vasquez, A. J. Easton, J. B. Domachowske, and H. F. Rosenberg. 2003. Diminished expression of an antiviral ribonuclease in response to pneumovirus infection in vivo. Antiviral Res. 59:181-191. [DOI] [PubMed] [Google Scholar]
  • 266.Munir, S., and V. Kapur. 2003. Regulation of host cell transcriptional physiology by the avian pneumovirus provides key insights into host-pathogen interactions. J. Virol. 77:4899-4910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Murphy, B. R., S. L. Hall, A. B. Kulkarni, J. E. Crowe, P. L. Collins, M. Connors, R. A. Karron, and R. M. Chanock. 1994. An update on the approaches to the development of respiratory syncytial virus (RSV) and parainfluenza virus type 3 (PIV3) vaccines. Virus Res. 32:13-36. [DOI] [PubMed] [Google Scholar]
  • 268.Murphy, B. R., and P. L. Collins. 2002. Live-attenuated virus vaccines for respiratory syncytial and parainfluenza viruses: applications of reverse genetics. J. Clin. Investig. 10:21-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Murphy, P. M. 1993. Molecular mimicry and the generation of host defense protein diversity. Cell 72:823-826. [DOI] [PubMed] [Google Scholar]
  • 270.Nakato, H., and K. Kimata. 2002. Heparan sulfate fine structure and specificity of proteoglycan functions. Biochim. Biophys. Acta 1573:312-318. [DOI] [PubMed] [Google Scholar]
  • 271.Naylor, C. J., K. J. Worthington, and R. C. Jones. 1997. Failure of maternal antibodies to protect young turkey poults against challenge with turkey rhinotracheitis virus. Avian Dis. 41:968-971. [PubMed] [Google Scholar]
  • 272.Nikitenko, A. A., Y. E. Raifeld, and T. Z. Wang. 2001. The discovery of RFI- 641 as a potent and selective inhibitor of the respiratory syncytial virus. Bioorg. Med. Chem. Lett. 11:1041-1044. [DOI] [PubMed] [Google Scholar]
  • 273.Nissen, M. D., I. M. Mackay, S. J. Withers, D. J. Sibert, and T. P. Sloots. 2002. Evidence of human metapneumovirus in Australian children. Med. J. Aust. 176:188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Njenga, M. K., H. M. Lwamba, and B. S. Seal. 2003. Metapneumoviruses in birds and humans. Virus Res. 91:163-169. [DOI] [PubMed] [Google Scholar]
  • 275.Olmsted, R. A., and P. L. Collins. 1989. The 1A protein of respiratory syncytial virus is an integral membrane protein present as multiple, structurally distinct species. J. Virol. 63:2019-2029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Ostler, T., W. Davidson, and S. Ehl. 2002. Virus clearance and immunopathology by CD8+ T cells during infection with respiratory syncytial virus are mediated by IFN-gamma. Eur. J. Immunol. 32:2117-2123. [DOI] [PubMed] [Google Scholar]
  • 277.Panigrahy, B., D. A. Senne, J. C. Pedersen, T. Gidlewski, and R. K. Edson. 2000. Experimental and serological observations on avian pneumovirus (APV/turkey/Colorado/97) infection in turkeys. Avian Dis. 44:17-22. [PubMed] [Google Scholar]
  • 278.Panuska, J. R., M. I. Hertz, H. Taraf, A. Villani, N. M. Cirino. 1992. Respiratory syncytial virus infection of alveolar macrophages in adult transplant patients. Am. Rev. Respir. Dis. 145:934-939. [DOI] [PubMed] [Google Scholar]
  • 279.Paradiso, P. R., S. W. Hildreth, D. A. Hogerman, D. J. Speelman, J. Oren, and D. H. Smith. 1994. Safety and immunogenicity of a subunit respiratory syncytial virus vaccine in children 24-48 months old. Pediatr. Infect. Dis. J. 13:792-798. [DOI] [PubMed] [Google Scholar]
  • 280.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]
  • 281.Pastey, M. K., J. E. Crowe, Jr., and B. S. Graham. 1999. RhoA interacts with the fusion glycoprotein of respiratory syncytial virus and facilitates virus-induced syncytium formation. J. Virol. 73:7262-7269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Pastey, M. K., T. L. Gower, P. W. Spearman, J. E. Crowe, Jr., and B. S. Graham. 2000. A RhoA-derived peptide inhibits syncytium formation induced by respiratory syncytial virus and parainfluenza virus type 3. Nat. Med. 6:35-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Patnayak, D. P., A. M. Sheikh, B. R. Bulati, and S. M. Goyal. 2002. Experimental and field evaluation of a live vaccine against avian pneumovirus. Avian Pathol. 31:377-382. [DOI] [PubMed] [Google Scholar]
  • 284.Peeples, M. E., and S. Levine. 1997. Respiratory syncytial virus polypeptides: their location in the virion. Virology 95:137-145. [DOI] [PubMed] [Google Scholar]
  • 285.Peeples, M. E., and P. L. Collins. 2000. Mutations in the 5′ trailer region of a respiratory syncytial virus minigenome which limit RNA replication to one step. J. Virol. 74:146-155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Peret, T. C., G. Boivin, Y. Li, M. Couillard, C. Humphrey, A. D. Osterhaus, D. D. Erdman, and L. J. Anderson. 2002. Characterization of human metapneumoviruses isolated from patients in North America. J. Infect. Dis. 185:1660-1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Philippou, S., P. Otto, P. Reinhold, M. Elschner, and H. J. Streckert. 2000. Respiratory syncytial virus-induced chronic bronchiolitis in experimentally-infected calves. Virchows Arch. 436:617-621. [DOI] [PubMed] [Google Scholar]
  • 288.Piedra, P. A., S. Grace, A. Jewell, S. Spinelli, D. Bunting, D. A. Hogerman, F. Malinoski, and P. W. Hiatt. 1996. Purified fusion protein vaccine protects against lower respiratory tract illness during respiratory syncytial virus season in children with cystic fibrosis. Pediatr. Infect. Dis. J. 15:23-31. [DOI] [PubMed] [Google Scholar]
  • 289.Poch, O., B. M. Blumberg, L. Bougueleret, and N. Tordo. 1990. Sequence comparison of five polymerases (L proteins) of unsegmented negative-strand RNA viruses: theoretical assignment of functional domains. J. Gen. Virol. 71:1153-1162. [DOI] [PubMed] [Google Scholar]
  • 290.Poch, O., L. Sauvaget, M. Delarue, and N. Tordo. 1989. Identification of four conserved motifs among the RNA-dependent polymerase encoding proteins. EMBO J. 8:3867-3874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.PREVENT Study Group. 1997. Reduction of respiratory syncytial virus hospitalization among premature infants and infant with bronchopulmonary dysplasia using respiratory syncytial virus immune globulin prophylaxis. Pediatrics 99:93-99. [DOI] [PubMed] [Google Scholar]
  • 292.Prince, G. A., A. Mathews, S. J. Curtis, and D. D. Porter. 2000. Treatment of respiratory syncytial virus bronchiolitis and pneumonia in a cotton rat model with systemically administered monoclonal antibody (Palivizumab) and glucocorticoid. J. Infect. Dis. 182:1326-1330. [Google Scholar]
  • 293.Pringle, C. R., and A. J. Easton. 1997. Monopartite negative strand RNA genomes. Semin. Virol. 8:49-57. [Google Scholar]
  • 294.Prober, C. G., and E. E. L. Wang. 1997. Reducing the morbidity of lower respiratory tract infections caused by respiratory syncytial virus: still no answer. Pediatrics 99:472-475. [DOI] [PubMed] [Google Scholar]
  • 295.Randhawa, J. S., P. Chambers, C. R. Pringle, and A. J. Easton. 1995. Nucleotide sequences of the genes encoding the putative attachment glycoprotein (G) of mouse and tissue culture-passaged strains of pneumonia virus of mice. Virology 207:240-245. [DOI] [PubMed] [Google Scholar]
  • 296.Randhawa, J. S., A. C. Marriott, C. R. Pringle, and A. J. Easton. 1997. Rescue of synthetic minireplicons establishes the absence of the NS1 and NS2 genes from avian pneumovirus. J. Virol. 71:9849-9854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Randhawa, J. S., S. D. Wilson, K. P. Tolley, D. Cavanagh, C. R. Pringle, and A. J. Easton. 1996. Nucleotide sequence of the gene encoding the viral polymerase of avian pneumovirus. J. Gen. Virol. 77:3047-3051. [DOI] [PubMed] [Google Scholar]
  • 298.Randolph, A. G., and E. E. L. Wang. 1996. Ribavirin for respiratory syncytial virus lower respiratory tract infection: a systematic overview. Arch. Pediatr. Adolesc. Med. 150:942-947. [DOI] [PubMed] [Google Scholar]
  • 299.Rautenschlein, S., A. M. Sheikh, D. P. Patnayak, R. L. Miller, J. M. Sharma, and S. M. Goyal. 2002. Effect of an immunomodulator on the efficacy of an attenuated vaccine against avian pneumovirus in turkeys. Avian Dis. 46:555-561. [DOI] [PubMed] [Google Scholar]
  • 300.Rawlinson, W. D., Z. Waliuzzaman, I. W. Carter, Y. C. Belessis, K. M. Gilbert, and J. R. Morton. 2003. Asthma exacerbations in children associated with rhinovirus but not human metapneumovirus infection. J. Infect. Dis. 187:1314-1318. [DOI] [PubMed] [Google Scholar]
  • 301.Raza, M. W., C. C. Blackwell, R. A. Elton, and D. M. Weir. 2000. Bactericidal activity of a monocytic cell line (THP-1) against common respiratory tract bacterial pathogens is depressed after infection with respiratory syncytial virus. J. Med. Microbiol. 49:227-233 [DOI] [PubMed] [Google Scholar]
  • 302.Reinhold, P., G. Becher, and M. Rothe. 2000. Evaluation of the measurement of leukotriene B4 concentration in exhaled condensate as a noninvasive method for assessing mediators of inflammation in the lungs of calves. Am. J. Vet. Res. 61:742-749. [DOI] [PubMed] [Google Scholar]
  • 303.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]
  • 304.Roberts, S. R., D. Lichtenstein, L. A. Ball, and G. W. Wertz. 1994. The membrane-associated and secreted forms of the respiratory syncytial virus attachment glycoprotein G are synthesized from alternative initiation codons. J. Virol. 68:4538-4546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Rodriguez, W. J., H. W. Kim, and C. D. Brandt. 1987. Aerosolized ribavirin in the treatment of patients with respiratory syncytial virus disease. Pediatr. Infect. Dis. J. 6:159-163. [DOI] [PubMed] [Google Scholar]
  • 306.Rodriguez, W. J., W. C. Gruber, J. R. Groothuis, E. A. Simoes, A. J. Rosas, M. Lepow, A. Kramer, and V. Hemming. 1997. Respiratory syncytial virus immune globulin treatment of RSV lower respiratory tract infection in previously healthy children. Pediatrics 100:937-942. [DOI] [PubMed] [Google Scholar]
  • 307.Roosevelt, G., K. Sheehan, J. Grupp-Phelan, R. R. Tanz, and R. Listernick. 1996. Dexamethasone in bronchiolitis: a randomised, controlled trial. Lancet 348:292-295. [DOI] [PubMed] [Google Scholar]
  • 308.Rontved, C. M., K. Tjornejoj, B. Viuff, L. E. Larsen, K. L. Godson, L. Roonsholt, and S. Alexandersen. 2000. Increased pulmonary secretion of tumor necrosis factor-alpha in calves experimentally infected with bovine respiratory syncytial virus. Vet. Immunol. Immunopathol. 31:199-214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Rosenberg, H. F., and J. I. Gallin. 2003. Inflammation, p. 1151-1170. In W. E. Paul (ed.), Fundamental immunology, 5th ed. Raven Press Ltd., New York, N.Y.
  • 310.Rosner, I., R. C. Welliver, P. Edelson, K. Geraci-Ciardullo, and M. Sun. 1987. Effect of ribavirin therapy on RSV-specific IgE and IgA responses after infection. J. Infect. Dis. 155:1043-1047. [DOI] [PubMed] [Google Scholar]
  • 311.Sakurai, H., R. A. Williamson, J. E. Crowe, J. A. Beeler, P. Poignard, R. B. Bastidas, R. M. Chanock, and D. R. Burton. 1999. Human antibody responses to mature and immature forms of viral envelope in respiratory syncytial virus infection: significance for subunit vaccines. J. Virol. 73:2956-2962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Samal, S. K., and P. L. Collins. 1996. RNA replication by a respiratory syncytial virus RNA analog does not obey the rule of six and retains a nonviral trinucleotide extension at the leader end. J. Virol. 70:5075-5082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 313.Sarmiento, R. E., R. Tirado, and B. Gomez. 2002. Characteristics of a respiratory syncytial virus persistently infected macrophage-like culture. Virus Res. 20:45-58. [DOI] [PubMed] [Google Scholar]
  • 314.Satake, M., N. Elango, and S. Venkatesan. 1984. Sequence analysis of the respiratory syncytial virus phosphoprotein gene. J. Virol. 52:991-994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Schlender, J., B. Bossert, U. Buchholz, and K. K. Conzelmann. 2000. Bovine respiratory syncytial virus nonstructural proteins NS1 and NS2 cooperatively antagonize alpha/beta interferon-induced antiviral response. J. Virol. 74:8234-8242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Schlender, J., G. Zimmer, G. Herrler, and K. K. Conzelmann. 2003. Respiratory syncytial virus (RSV) fusion protein subunit F2, not attachment protein G, determines specificity of RSV infection. J. Virol. 77:4609-4616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Schmidt, U., J. Beyer, U. Polster, L. J. Gershwin, and U. J. Buchholz. 2002. Mucosal immunization with live recombinant bovine respiratory syncytial virus (BRSV) and recombinant BRSV lacking the envelope glycoprotein G protects against challenge with wild-type BRSV. J. Virol. 76:12355-12359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Schreiber, P., J. P. Matheise, F. Dessy, M. Heimann, J. J. Letesson, P. Coppe, and A. Collard. 2000. High mortality rate associated with bovine respiratory syncytial virus (BRSV) infection in Belgian white blue calves previously vaccinated with an inactivated BRSV vaccine. J. Vet. Med. Ser. B. 47:535-550. [DOI] [PubMed] [Google Scholar]
  • 319.Schwarze, J., G. Ciesiewicz, A. Joetham, T. Ikemura, M. J. Makela, A. Dakhar, L. D. Shultz, M. C. Lamers, and E. W. Gelfand. 2000. Critical roles for interleukin-4 and interleukin-5 during respiratory syncytial virus infection in the development of airway hyperresonsiveness after airway sensitiziation. Am. J. Respir. Crit. Care Med. 162:380-386. [DOI] [PubMed] [Google Scholar]
  • 320.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 airways hyperresonsiveness following acute respiratory syncytial virus infection. J. Immunol. 162:2997-3004. [PubMed] [Google Scholar]
  • 321.Shin, H. J., M. K. Njenga, B. McComb, D. A. Halvorson, and K. V. Nagaraja. 2000. Avian pneumovirus (APV) RNA from wild and sentinel birds in the United States has genetic homology with RNA from APV isolates from domestic turkeys. J. Clin. Microbiol. 38:4282-4284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322.Shin, H. J., G. Rajashekara, F. F. Jirjis, D. P. Shaw, S. M. Goyal, D. A. Halvorson, and K. V. Nagaraja. 2000. Specific detection of avian pneumovirus (APV) US isolates by RT-PCR. Arch. Virol. 145:1239-1246. [DOI] [PubMed] [Google Scholar]
  • 323.Siber, G. R., R. D. Leombruno, and J. Leszczynski. 1994. Comparison of antibody concentrations and protective activity of respiratory syncytial virus immune globulin and conventional immune globulin. J. Infect. Dis. 169:1368-1373. [DOI] [PubMed] [Google Scholar]
  • 324.Simoes, E. A. F., H. M. Sondheimer, and H. C. Meissner. 1996. Respiratory syncytial virus immunoglobulin as prophylaxis against respiratory syncytial virus in children with congenital heart disease. Pediatr. Res. 662:113A. [DOI] [PubMed] [Google Scholar]
  • 325.Simoes, E. A., and J. R. Groothuis. 2002. Respiratory syncytial virus prophylaxis—the story so far. Respir. Med. 96:S15-S24. [DOI] [PubMed] [Google Scholar]
  • 326.Slack, M. S., and A. J. Easton. 1998. Characterisation of the interaction of the human respiratory syncytial virus phosphoprotein and nucleocapsid protein using the two-hybrid system. Virus Res. 55:167-176. [DOI] [PubMed] [Google Scholar]
  • 327.Smith, D. W., L. R. Frankel, L. H. Mathers, A. T. S. Tang, R. L. Ariaggno, and C. G. Prober. 1991. A controlled trial of aerosolized ribavirin in infants receiving mechanical ventilation for severe respiratory syncytial virus infection. N. Engl. J. Med. 325:24-29. [DOI] [PubMed] [Google Scholar]
  • 328.Smith, P. K., S. Z. Wang, K. D. Dowling, and K. D. Forsyth. 2001. Leucocyte populations in respiratory syncytial virus-induced bronchiolitis J. Paediatr. Child Health 37:146-151 [DOI] [PubMed] [Google Scholar]
  • 329.Spann, K. M., P. L. Collins, and M. N. Teng. 2003. Genetic recombination during coinfection of two mutants of human respiratory syncytial virus. J. Virol. 77:11201-11211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Spillmann, D. 2001. Heparan sulfate: anchor for viral intruders? Biochimie 83:811-817. [DOI] [PubMed] [Google Scholar]
  • 331.Springer, C., E. Bar-Yishay, K. Uwayyed, A. Avital, D. Vilozni, and S. Godfrey. 1990. Corticosteroids do not affect the clinical or physiological status of infants with bronchiolitis. Pediatr. Pulmonol. 9:181-185. [DOI] [PubMed] [Google Scholar]
  • 332.Stec, D. S., M. G. Hill III, and P. L. Collins. 1991. Sequence analysis of the polymerase L gene of human respiratory syncytial virus and predicted phylogeny of nonsegmented negative-strand viruses. Virology 183:273-287. [DOI] [PubMed] [Google Scholar]
  • 333.Sudo, K., K. Konno, W. Watanabe, S. Shigeta, and T. Yokota. 2001. Mechanism of selective inhibition of respiratory syncytial virus by a benzodithiin compound (RD3-0028). Microbiol. Immunol. 45:531-537. [DOI] [PubMed] [Google Scholar]
  • 334.Sussman, S., M. Grossman, R. Magoffin, and J. Schieble. 1964. Dexamethasone (16-alpha, 9-alpha-fluoroprednisolone) in obstructive respiratory tract infections in children. Pediatrics 34:851-855. [PubMed] [Google Scholar]
  • 335.Sutherland, K. A., P. L. Collins, and M. E. Peeples. 2001. Synergistic effects of gene-end signal mutations and the M2-1 protein on transcription termination by respiratory syncytial virus. Virology 288:295-307. [DOI] [PubMed] [Google Scholar]
  • 336.Taber, L. H., V. Knight, and B. E. Gilbert. 1983. Ribavirin aerosol treatment of bronchiolitis associated with respiratory syncytial virus infection in infants. Pediatrics 72:613-618. [PubMed] [Google Scholar]
  • 337.Takao, S., H. Shimozono, H. Kashiwa, Y. Shimazu, S. Fukuda, M. Kuwayama, and K. Miyazaki. 2003. Clinical study of pediatric cases of acute respiratory diseases associated with human metapneumovirus in Japan. Jpn. J. Infect. Dis. 56:127-129. [PubMed] [Google Scholar]
  • 338.Tal, A., C. Bavilski, and D. Yohai. 1983. Dexamethasone and salbutamol in the treatment of acute wheezing in infants. Pediatrics 71:13-18. [PubMed] [Google Scholar]
  • 339.Tang, R. S., N. Nguyen, H. Zhou, and H. Jin. 2002. Clutsered charge-to-alanine mutagenesis of human respiratory syncytial virus L polymerase generates temperature-sensitive viruses. Virology 302:207-216. [DOI] [PubMed] [Google Scholar]
  • 340.Taylor, G. S., I. B. Vipond, and E. O. Caul. 2001. Molecular epidemiology of outbreak of respiratory syncytial virus within bone marrow transplantation unit. J. Clin. Microbiol. 39:801-803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Techaarpornkul, S., N. Barretto, and M. E. Peeples. 2001. Functional analysis of recombinant respiratory syncytial virus deletion mutants lacking the small hydrophobic and/or attachment glycoprotein gene. J. Virol. 75:6825-6834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Techaarpornkul, S., P. L. Collins, and M. E. Peeples. 2002. Respiratory syncytial virus with the fusion protein as its only viral glycoprotein is less dependent on cellular glycosaminoglycans for attachment than complete virus. Virology 294:296-304. [DOI] [PubMed] [Google Scholar]
  • 343.Teng, M. N., and P. L. Collins. 1998. Identification of the respiratory syncytial virus proteins required for formation and passage of helper-dependent infectious particles. J. Virol. 72:5707-5716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Teng, M. N., and P. L. Collins. 1999. Altered growth characteristics of recombinant respiratory syncytial viruses which do not produce NS2 protein. J. Virol. 73:466-473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Teng, M. N., and P. L. Collins. 2002. The central conserved cystine noose of the attachment G protein of human respiratory syncytial virus is not required for efficient viral infection in vitro or in vivo. J. Virol. 76:6164-6171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Teng, M. N., S. S. Whitehead, A. Bermingham, M. St. Claire, W. R. Elkins, B. R. Murphy, and P. L. Collins. 2000. Recombinant respiratory syncytial virus that does not express the NS1 or M2-2 protein is highly attenuated and immunogenic in chimpanzees. J. Virol. 74:9317-9321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Teng, M. N., S. S. Whitehead, and P. L. Collins. 2001. Contribution of the respiratory syncytial virus G glycoprotein and its secreted and membrane-bound forms to virus replication in vitro and in vivo. Virology 289:283-296. [DOI] [PubMed] [Google Scholar]
  • 348.Reference deleted.
  • 349.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 netword\ks in respiratory syncytial virus-infected cells. J. Virol. 76:6800-6814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Tjornehoj, K., A. Uttenthal, B. Viuff, L. E. Larsen, C. Rontved, and L. Ronsholt. 2003. An experimental infection model for reproduction of calf pneumonia with bovine respiratory syncytial virus (BRSV) based on one combined exposure of calves. Res. Vet. Sci. 74:55-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Tolley, K. P., A. C. Marriott, A. Simpson, D. J. Plows, D. A. Matthews, S. J. Longhurst, J. E. Evans, J. L. Johnson, P. A. Cane, V. B. Randolph, A. J. Easton, and C. R. Pringle. 1996. Identification of mutations contributing to the reduced virulence of a modified strain of respiratory syncytial virus. Vaccine 14:1637-1646. [DOI] [PubMed] [Google Scholar]
  • 352.Tripp, R. A., L. Jones, and L. J. Anderson. 2000. Respiratory syncytial virus G and/or SH glycoproteins modify CC and CXC chemokine mRNA expression in the BALB/c mouse. J. Virol. 74:6227-6229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Tripp, R. A., L. P. Jones, L. M. Haynes, H. Zheng, P. M. Murphy, and L. J. Anderson. 2001. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nat. Immunol. 2:732-738. [DOI] [PubMed] [Google Scholar]
  • 354.Tristram, D. A., R. C. Welliver, C. K., Mohar, D. A. Hogeman, S. W. Hildreth, and P. Paradiso. 1993. Immunogenicity and safety of respiratory syncytial virus subunit vaccine in seropositive children 18-36 months old. J. Infect. Dis. 167:191-195. [DOI] [PubMed] [Google Scholar]
  • 355.Trudel, M., F. Nadon, C. Simard, F. Belanger, R. Alain, C. Seguin, and G. Lussier. 1989. Comparison of caprine, human and bovine strains of respiratory syncytial virus. Arch. Virol. 107:141-149. [DOI] [PubMed] [Google Scholar]
  • 356.Tsutsumi, H., R. Takeuchi, M. Ohskai, K. Seki, and S. Chiba. 1999. Respiratory syncytial virus infection of human respiratory epithelial cells enhances inducible nitric oxide synthase gene expression. J. Leukoc. Biol. 66:99-104. [PubMed] [Google Scholar]
  • 357.Valarcher, J. F., F. Schelcher, and H. Bourhy. 2000. Evolution of bovine respiratory syncytial virus. J. Virol. 74:10714-10728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Valarcher, J. F., J. Furze, S. Wyld, R. Cook, K. K. Conzelmann, and G. Taylor. 2003. Role of alpha/beta interferons in the attenuation and immunogenicity of recombinant bovine respiratory syncytial viruses lacking NS proteins. J. Virol. 77:8426-8439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.van Aerschot, A. A., P. Mamos, N.J. Weyns, S. Ikeda, E. DeClercq, and P. A. Herdewijn. 1993. Antiviral activity of C-alkylated purine nucleosides obtained by cross-coupling with tetraalkylting reagent. J. Med. Chem. 36:2938-2942. [DOI] [PubMed] [Google Scholar]
  • 360.van den Hoogen, B. G., J. C. de Jong, J. Groen, T. Kuiken, R. de Groot, R. A. M. Foucher, and A. D. M. E. Osterhaus. 2001. A newly discovered human pneumovirus isolated from young children with respiratory tract disease. Nat. Med. 7:719-724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361.van den Hoogen, B. G., T. M. Bestebroer, A. D. Osterhaus, and R. A. Fouchier 2002. Analysis of the genomic sequence of a human metapneumovirus. Virology 295:119-132. [DOI] [PubMed] [Google Scholar]
  • 362.van der Sluijs, K. F., L. Van Elden, M. Nijhuis, R. Schuurman, S. Florquin, H. M. Jansen, R. Lutter, and T. Van der Poll. 2003. Toll-like receptor 4 is not involved in host defense against respiratory tract infection with Sendai virus. Immunol. Lett. 89:201-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.van de Zande, S., H. Nauwynchk, and M. Pensaert. 2002. Efficacy of avian pneumovirus vaccines against an avian pneumovirus/Escherichia coli O2:K1 dual infection in turkeys. Vet. Rec. 150:340-343. [DOI] [PubMed] [Google Scholar]
  • 364.van Schaik, S. M., N. Obot, G. Enhorning, K. Hintz, K. Gross, G. E. Hancock, A. M. Stack, and R. C. Welliver. 2000. Role of interferon gamma in the pathogenesis of primary respiratory syncytial virus infection in BALB/c mice. J. Med. Virol. 62:257-266. [DOI] [PubMed] [Google Scholar]
  • 365.van Woensel, J. B., T. F. Wolfs, W. M. van Aalderen, P. L. Brand, and J. L. Kimpen. 1997. Randomised double blind placebo controlled trial of prednisolone in children admitted to hospital with respiratory syncytial virus bronchiolitis. Thorax 52:634-637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Villanueva, N., R. Hardy, A. Asenjo, Q. Yu, and G. Wertz. 2000. The bulk of the phosphorylation of human respiratory syncytial virus phosphoprotein is not essential but modulates viral RNA transcription and replication. J. Gen Virol. 81:129-133. [DOI] [PubMed] [Google Scholar]
  • 367.Viuff, B., K. Tjornehoj, L. E. Larsen, C. M. Rontved, A. Uttenthal, L. Ronsholt, and S. Alexandersen. 2002. Replication and clearance of respiratory syncytial virus: apoptosis is an important pathway of virus clearance after experimental infection with bovine respiratory syncytial virus. Am. J. Pathol. 161:2195-2207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Walsh, E. E., A. R. Falsey, and W. M. Sullender. 1998. Monoclonal antibody neutralization escape mutants of respiratory syncytial virus with unique alterations in the attachment (G) protein. J. Gen. Virol. 79:479-487. [DOI] [PubMed] [Google Scholar]
  • 369.Wang, E. E. L., B. J. Law, and F. D. Boucher. 1996. Pediatric investigators collaborative network in infections in Canada (PICNIC) study of admission and management variation in patients hospitalized with respiratory syncytial lower respiratory tract infection. J. Pediatr. 129:390-395. [DOI] [PubMed] [Google Scholar]
  • 370.Wang, S. Z., and K. D. Forsyth. 2000. The interaction of neutrophils with respiratory epithelial cells in viral infection. Respirology 5:1-10. [DOI] [PubMed] [Google Scholar]
  • 371.Weisman, L. E. 2003. Populations at risk for developing respiratory syncytial virus and risk factors for respiratory syncytial virus severity: infants with predisposing conditions. Pediatr. Infect. Dis. J. 22:S33-S37. [DOI] [PubMed] [Google Scholar]
  • 372.Welliver, R. C. 2003. Respiratory syncytial virus and other respiratory viruses. Pediatr. Infect. Dis. J. 22:S6-S12. [DOI] [PubMed] [Google Scholar]
  • 373.Wertz, G. W., P. L. Collins, Y. Huang, C. Gruber, S. Levine, and L. A. Ball. 1985. Nucleotide sequence of the G protein gene of human respiratory syncytial virus reveals an unusual type of viral membrane protein. Proc. Natl. Acad. Sci. USA 82:4075-4079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.West, K., L. Petrie, C. Konoby, D. M. Haines, V. Cortese, and J. A. Ellis. 2000. The efficacy of modified-live bovine respiratory syncytial virus vaccines in experimentally infected calves. Vaccine 18:907-919. [DOI] [PubMed] [Google Scholar]
  • 375.West, K., L. Petrie, D. M. Haines, C. Konoby, E. G. Clark, K. Martin, and J. A. Ellis. 1999. The effect of formalin-inactivated vaccine on respiratory disease associated with bovine respiratory syncytial virus infection in calves. Vaccine 17:809-820. [DOI] [PubMed] [Google Scholar]
  • 376.Wheeler, J. G., J. Wofford, and R. B. Turner. 1993. Historical cohort evaluation of ribavirin efficacy in respiratory syncytial virus infection. Pediatr. Infect. Dis. J. 12:209-213. [DOI] [PubMed] [Google Scholar]
  • 377.Whimbey, E., R. E. Champlin, J. A. Englund, N. Q. Mirza, P. A. Piedra, J. M. Goodrich, D. Przepiorka, M. A. Luna, R. C. Morice, and J. L. Neumann. 1995. Combination therapy with aerosolized ribavirin and intravenous immunoglobulin for respiratory syncytial virus disease in adult bone marrow transplant recipients. Bone Marrow Transplant. 16:393-399. [PubMed] [Google Scholar]
  • 378.Whitehead, S. S., A. Bukreyev, M. N. Teng, C. Y. Firestone, M. St. Claire, W. R. Elkins, P. L. Collins, and B. R. Murphy. 1999. Recombinant respiratory syncytial virus bearing a deletion of either the NS2 or SH gene is attenuated in chimpanzees. J. Virol. 73:3438-3442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379.Woelk, C. H., and E. C. Holmes. 2001. Variable immune-driven natural selection in the attachment (G) glycoprotein of respiratory syncytial virus (RSV). J. Mol. Evol. 52:182-192. [DOI] [PubMed] [Google Scholar]
  • 380.Woolums, A. R., M. L. Anderson, R. A. Gunther, E. S. Schelegle, D. R. LaRochelle, R. S. Singer, G. A. Boyle, K. E. Friebertshauser, and L. J. Gershwin. 1999. Evaluation of severe disease induced by aerosol inoculation of calves with bovine respiratory syncytial virus. Am. J. Vet. Res. 60:473-480. [PubMed] [Google Scholar]
  • 381.Worthington, K. J., B. A. Sargent, F. G. Davelaar, and R. C. Jones. 2003. Immunity to avian pneumovirus infection in turkeys following in ovo vaccination with an attenuated vaccine. Vaccine 21:1355-1362. [DOI] [PubMed] [Google Scholar]
  • 382.Wunner, W. H., and C. R. Pringle. 1976. Respiratory syncytial virus proteins. Virology 73:228-243. [DOI] [PubMed] [Google Scholar]
  • 383.Wyde, P. R., D. K. Moore, D. M. Pimentel, and H. A. Blough. 1995. Evaluation of the antiviral activity of N-(phosphonoacetyl)-l-aspartate against paramyxoviruses in tissue culture and against respiratory syncytial virus in cotton rats. Antiviral Res. 27:59-69. [DOI] [PubMed] [Google Scholar]
  • 384.Wyde, P. R., D. K. Moore, D. M. Pimentel, B. E. Gilbert, R. Nimrod, and A. Panet. 1996. Recombinant superoxide dismutase (SOD) administered by aerosol inhibits respiratory syncytial virus infection in cotton rats. Antiviral Res. 31:173-184. [DOI] [PubMed] [Google Scholar]
  • 385.Young, D. F., L. Didcock, S. Goodbourn, and R. E. Randall. 2000. Paramyxoviridae use distinct virus-specific mechanisms to circumvent the interferon response. Virology 269:383-390. [DOI] [PubMed] [Google Scholar]
  • 386.Yu, Q., P. J. Davis, T. Barrett, M. M. Binns, M. E. Boursnell, and D. Cavanagh. 1991. Deduced amino acid sequence of the fusion glycoprotein of turkey rhinotracheitis virus has greater identity with that of human respiratory syncytial virus, a pneumovirus, than that of paramyxoviruses and morbilliviruses. J. Gen. Virol. 72:75-81. [DOI] [PubMed] [Google Scholar]
  • 387.Yu, Q., P. J. Davis, T. D. Brown, and D. Cavanagh. 1992. Sequence and in vitro expression of the M2 gene of turkey rhinotracheitis pneumovirus. J. Gen. Virol. 73:1355-1363. [DOI] [PubMed] [Google Scholar]
  • 388.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 RSV 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]
  • 389.Yunus, A. S., S. K. Khattar, P. L. Collins, and S. K. Samal. 2001. Rescue of bovine respiratory syncytial virus from cloned cDNA: entire genome sequence of BRSV strain A51908. Virus Genes 23:157-164. [DOI] [PubMed] [Google Scholar]
  • 390.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]
  • 391.Zhang, Y., M. Jamaluddin, S. Wang, B. Tian, R. Garofalo, A. Casola, and A. R. Brasier. 2003. Ribavirin treatment up-regulates antiviral gene expression via the interferon-stimulated response element in respiratory syncytial virus-infected epithelial cells. J. Virol. 77:5933-5947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Zimmer, G., K. K. Conzelmann, and G. Herrler. 2002. Cleavage at the furin consensus sequence RAR/KR(109) and presence of the intervening peptide of the respiratory syncytial virus fusion protein are dispensable for virus replication in cell culture. J. Virol. 76:9218-9224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.Zimmer, G., L. Budz, and G. Herrler. 2001. Proteolytic activation of respiratory syncytial virus fusion protein. Cleavage at two furin consensus sequences. J. Biol. Chem. 276:31642-31650. [DOI] [PubMed] [Google Scholar]
  • 394.Zimmer, G., M. Rohn, G. P. McGregor, M. Schemann, K. K. Conzelmann, and G. Herrler. 2003. Virokinin, a bioactive peptide of the tachykinin family, is released from the fusion protein of bovine respiratory syncytial virus. J. Biol. Chem. 278:46854-46861. [DOI] [PubMed] [Google Scholar]

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

RESOURCES